1
|
Mageswaran N, Zainal SH, Hassan NI, Abd Karim NH, Ismail NAS. Emerging Biomarkers and Electrochemical Biosensors for Early Detection of Premature Coronary Artery Disease. Diagnostics (Basel) 2025; 15:940. [PMID: 40218291 PMCID: PMC11988804 DOI: 10.3390/diagnostics15070940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025] Open
Abstract
Coronary artery disease (CAD) is one of the primary causes of morbidity and death worldwide. Premature CAD (pCAD) is the term used to describe the 3-10% of CAD occurrences that occur in people under 45 worldwide. Diagnostic difficulties arise from the different risk factor profiles of pCAD and late-onset CAD. Better cardiovascular risk prediction in younger populations has been made possible by the development of biomarker detection tools. This can be applied to a diagnostic tool, including electrochemical biosensors, which have been predicted to be instrumental because of their adaptability for point-of-care applications for quicker diagnoses. These biosensors provide efficient, scalable, and reasonably priced solutions for the quick identification and tracking of CAD. Multiplex biomarker detection has been adopted as a viable approach for early diagnosis and risk assessment due to the constraints of using a single biomarker for pCAD diagnosis. Thus, this study looks at current developments in biosensing technology and discusses established and new cardiac biomarker panels for pCAD identification.
Collapse
Affiliation(s)
- Nanthini Mageswaran
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Sarah Husnaini Zainal
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nurul Izzaty Hassan
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (N.I.H.); (N.H.A.K.)
| | - Nurul Huda Abd Karim
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (N.I.H.); (N.H.A.K.)
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
2
|
Fu X, Zhang X, Wang L, Zhang J, Li W, Qin S, Zhang M, Zheng X, Li Y, Yang S, Xue Q. The potential value of microRNA-409-5p-mediated negative regulation of USP7 in the diagnosis and treatment of acute myocardial infarction. BMC Cardiovasc Disord 2025; 25:167. [PMID: 40057674 PMCID: PMC11890518 DOI: 10.1186/s12872-025-04590-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
PURPOSE This study aimed to explore the effects of ubiquitin-specific peptidase 7 (USP7) on acute myocardial infarction (AMI) and the negative regulation of USP7 by microRNA-409-5p (miR-409-5p). METHODS Clinical data were collected from patients admitted to the Cardiology Department of Yan'an Hospital of Kunming City between July 2020 and July 2021. The participants included patients with AMI (AMI; n = 30), stable angina pectoris (SAP; n = 30), and chest pain syndrome (CPS; n = 30) and healthy controls (n = 30). The expression levels of miR-409-5p and USP7 were analysed using Quantitative real-time polymerase chain reaction (qRT‒PCR) and Western blotting (WB). Finally, a dual-luciferase assay was performed to verify the interaction between miR-409-5p and USP7. RESULTS The expression level of miR-409-5p was significantly lower (all p < 0.05), whereas the expression level of USP7 was elevated in patients with AMI compared with those in the other three groups (all p < 0.05). A dual-luciferase assay demonstrated that miR-409-5p binds to USP7 3'UTP to inhibit luciferase expression. Compared with cells transfected with mutation fragments and a luciferase reporter vector with microRNA-409-5p mimics and USP7 3'UTR binding and mutation sites, the luminescence level of cells with miR-409-5p was approximately 40% lower. Additionally, miRNA-409-5p was inversely correlated with cTnI (p = 0.004). CONCLUSION USP7 plays a significant role in AMI via negative regulation by miR-409-5p. Both miR-409-5p and USP7 hold key potential as early diagnostic biomarkers and therapeutic targets for AMI in the future.
Collapse
Affiliation(s)
- Xuemei Fu
- Department of Cardiology, Yan'an Hospital of Kunming City, Yan'an Hospital Affiliated with Kunming Medical University, Kunming, 650051, China
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Xi Zhang
- Department of Cardiology, Yan'an Hospital of Kunming City, Yan'an Hospital Affiliated with Kunming Medical University, Kunming, 650051, China
- Kunming Cardiovascular Interventional Imaging Institute, Kunming, 650051, China
| | - Lixing Wang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming, 650101, China
| | - Juan Zhang
- Department of Otolaryngology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Waiqiong Li
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Shaoxi Qin
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Min Zhang
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Xiaotian Zheng
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Ying Li
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China
| | - Shaobo Yang
- Department of Cardiology, Qujing Second People'S Hospital of Yunnan Province, Qujing, 655099, China.
| | - Qiang Xue
- Department of Cardiology, Yan'an Hospital of Kunming City, Yan'an Hospital Affiliated with Kunming Medical University, Kunming, 650051, China.
- Kunming Cardiovascular Interventional Imaging Institute, Kunming, 650051, China.
| |
Collapse
|
3
|
Ko TH, Kim Y, Jin C, Yu B, Lee M, Luong PK, Trinh TN, Yang Y, Kang H, Zhang Y, Ma R, Yoo K, Choi J, Kim JY, Woo SH, Han K, Choi JI. Shank3 Overexpression Leads to Cardiac Dysfunction in Mice by Disrupting Calcium Homeostasis in Cardiomyocytes. Korean Circ J 2025; 55:100-117. [PMID: 39506183 DOI: 10.4070/kcj.2024.0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVES SH3 and multiple ankyrin repeat domains 3 (Shank3) proteins play crucial roles as neuronal postsynaptic scaffolds. Alongside neuropsychiatric symptoms, individuals with SHANK3 mutations often exhibit symptoms related to dysfunctions in other organs, including the heart. However, detailed insights into the cardiac functions of Shank3 remain limited. This study aimed to characterize the cardiac phenotypes of Shank3-overexpressing transgenic mice and explore the underlying mechanisms. METHODS Cardiac histological analysis, electrocardiogram and echocardiogram recordings were conducted on Shank3-overexpressing transgenic mice. Electrophysiological properties, including action potentials and L-type Ca²⁺ channel (LTCC) currents, were measured in isolated cardiomyocytes. Ca²⁺ homeostasis was assessed by analyzing cytosolic Ca²⁺ transients and sarcoplasmic reticulum Ca²⁺ contents. Depolarization-induced cell shortening was examined in cardiomyocytes. Immunoprecipitation followed by mass spectrometry-based identification was employed to identify proteins in the cardiac Shank3 interactome. Western blot and immunocytochemical analyses were conducted to identify changes in protein expression in Shank3-overexpressing transgenic cardiomyocytes. RESULTS The hearts of Shank3-overexpressing transgenic mice displayed reduced weight and increased fibrosis. In vivo, sudden cardiac death, arrhythmia, and contractility impairments were identified. Shank3-overexpressing transgenic cardiomyocytes showed prolonged action potential duration and increased LTCC current density. Cytosolic Ca²⁺ transients were increased with prolonged decay time, while sarcoplasmic reticulum Ca²⁺ contents remained normal. Cell shortening was augmented in Shank3-overexpressing transgenic cardiomyocytes. The cardiac Shank3 interactome comprised 78 proteins with various functions. Troponin I levels were down-regulated in Shank3-overexpressing transgenic cardiomyocytes. CONCLUSIONS This study revealed cardiac dysfunction in Shank3-overexpressing transgenic mice, potentially attributed to changes in Ca²⁺ homeostasis and contraction, with a notable reduction in troponin I.
Collapse
Affiliation(s)
- Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
| | - Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Chunmei Jin
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Byeongil Yu
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Minju Lee
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Phuong Kim Luong
- Laboratory of Pathophysiology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Tran Nguyet Trinh
- Laboratory of Pathophysiology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Yeji Yang
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Kwangmin Yoo
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Korea
| | - Jungmin Choi
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, Korea
| | - Sun-Hee Woo
- Laboratory of Pathophysiology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea.
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
4
|
Celeski M, Segreti A, Crisci F, Cricco R, Piscione M, Di Gioia G, Nusca A, Fossati C, Pigozzi F, Ussia GP, Solaro RJ, Grigioni F. The Role of Cardiac Troponin and Other Emerging Biomarkers Among Athletes and Beyond: Underlying Mechanisms, Differential Diagnosis, and Guide for Interpretation. Biomolecules 2024; 14:1630. [PMID: 39766337 PMCID: PMC11727179 DOI: 10.3390/biom14121630] [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: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025] Open
Abstract
Cardiovascular (CV) disease remains the leading cause of morbidity and mortality worldwide, highlighting the necessity of understanding its underlying molecular and pathophysiological pathways. Conversely, physical activity (PA) and exercise are key strategies in reducing CV event risks. Detecting latent CV conditions in apparently healthy individuals, such as athletes, presents a unique challenge. The early identification and treatment of CV disorders are vital for long-term health and patient survival. Cardiac troponin is currently the most commonly used biomarker for assessing CV changes in both athletes and the general population. However, there remains considerable debate surrounding the mechanisms underlying exercise-induced troponin elevations and its release in non-ischemic contexts. Thus, there is a pressing need to identify and implement more sensitive and specific biomarkers for CV disorders in clinical practice. Indeed, research continues to explore reliable biomarkers for evaluating the health of athletes and the effectiveness of physical exercise. It is essential to analyze current evidence on troponin release in non-ischemic conditions, post-strenuous exercise, and the complex biological pathways that influence its detection. Furthermore, this study summarizes current research on cytokines and exosomes, including their physiological roles and their relevance in various CV conditions, especially in athletes. In addition, this paper gives special attention to underlying mechanisms, potential biomarkers, and future perspectives.
Collapse
Affiliation(s)
- Mihail Celeski
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Andrea Segreti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Filippo Crisci
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Riccardo Cricco
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Mariagrazia Piscione
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Giuseppe Di Gioia
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
- Institute of Sports Medicine and Science, Italian National Olympic Committee, Largo Piero Gabrielli 1, 00197 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Ross John Solaro
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
5
|
Wang N, Huang J, Fang Y, Du H, Chen Y, Zhao S. Molecular biomarkers of blunt cardiac injury: recent advances and future perspectives. Expert Rev Mol Diagn 2024; 24:1023-1031. [PMID: 39285529 DOI: 10.1080/14737159.2024.2405919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Blunt cardiac injury (BCI), associated with high morbidity and mortality, involves multiple injuries. With no widely accepted gold standard diagnostic test and molecular biomarkers still in debate and far from application in clinical practice, exploring specific molecular biomarkers of BCI is of great significance. The clarification of molecular biomarkers can improve the diagnosis of BCI, leading to more precise care for victims in various situations. AREAS COVERED Using the search term 'Biomarker AND Blunt cardiac injury,' we carefully reviewed related papers from June 2004 to June 2024 in PubMed and CNKI. After reviewing, we included 20 papers, summarizing the biomarkers reported in previous studies, and then reviewed molecular biomarkers such as troponins, Nterminal proBtype natriuretic peptide (NT proBNP), hearttype fatty acid binding protein (hFABP), and lactate for BCI diagnosis. Finally, valuable views on future research directions for diagnostic biomarkers of BCI were presented. EXPERT OPINION Several advanced technologies have been introduced into clinical medicine, which have ultimately changed the research on cardiac diseases in recent years. Some biomarkers have been identified and utilized for BCI diagnosis. Herein, we summarize the latest relevant information as a reference for clinical practice and future studies.
Collapse
Affiliation(s)
- Ning Wang
- Department of Forensic Pathology, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiliang Huang
- Department of Gastroenterology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Ying Fang
- Department of Forensic Pathology, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honglin Du
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanlin Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuquan Zhao
- Department of Forensic Pathology, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Jauhar MM, Damairetha FR, Mardliyati E, Ulum MF, Syaifie PH, Fahmi F, Satriawan A, Shalannanda W, Anshori I. Bioinformatics design of peptide binding to the human cardiac troponin I (cTnI) in biosensor development for myocardial infarction diagnosis. PLoS One 2024; 19:e0305770. [PMID: 39436888 PMCID: PMC11495608 DOI: 10.1371/journal.pone.0305770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/04/2024] [Indexed: 10/25/2024] Open
Abstract
Cardiovascular disease has reached a mortality rate of 470,000 patients each year. Myocardial infarction accounts for 49.2% of these deaths, and the cTnI protein is a crucial target in diagnosing myocardial infarction. A peptide-based bioreceptor design using a computational approach is a good candidate to be developed for a rapid, effective, and selective detection method for cTnI although it is still lacking in study. Hence, to address the scientific gap, we develop a new candidate peptide for the cTnI biosensor by bioinformatics method and present new computational approaches. The sequential point mutations were made to the selected peptide to increase its stability and affinity for cTnI. Next, molecular docking was performed to select the mutated peptide, and one of the best results was subjected to the molecular dynamics simulation. Finally, the results showed that the best peptide showed the lowest affinity and good stability among other mutated peptide designs for interacting with the cTnI protein. In addition, the peptide has been tested to have a higher specificity towards cTnI than its major isomer, sTnI, through molecular docking and molecular dynamics simulation. Therefore, the peptide is considered a good potential bioreceptor for diagnosing myocardial infarction diseases.
Collapse
Affiliation(s)
- Muhammad Miftah Jauhar
- COE Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
- Biomedical Engineering, Graduate School of Universitas Gadjah Mada, Sleman Regency, Special Region of Yogyakarta, Indonesia
| | - Filasta Rachel Damairetha
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, West Java, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), Cibinong, West Java, Indonesia
| | - Mokhamad Fakhrul Ulum
- School of Veterinary Medicine and Biomedical Sciences, IPB University (Bogor Agricultural University), Bogor, West Java, Indonesia
| | - Putri Hawa Syaifie
- COE Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
| | - Fahmi Fahmi
- Department of Electrical Engineering, Faculty of Engineering, Universitas Sumatera Utara, Medan, North Sumatera, Indonesia
| | - Ardianto Satriawan
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, West Java, Indonesia
| | - Wervyan Shalannanda
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, West Java, Indonesia
| | - Isa Anshori
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, West Java, Indonesia
- Center for Health and Sports Technology, Bandung Institute of Technology, Bandung, West Java, Indonesia
- Research Center for Nanosciences and Nanotechnology (RCNN), Bandung Institute of Technology, Bandung, West Java, Indonesia
| |
Collapse
|
7
|
Wang F, Chen F, Song W, Li Y, Wu H, Tian T, Tian M, Tang D, Liu Y. Sodium Fluoride Exposure Induces Developmental Toxicity and Cardiotoxicity in Zebrafish Embryos. Biol Trace Elem Res 2024:10.1007/s12011-024-04381-4. [PMID: 39287768 DOI: 10.1007/s12011-024-04381-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/14/2024] [Indexed: 09/19/2024]
Abstract
Fluorosis is a worldwide public health problem, in which the heart is an important target organ. However, studies on its toxicological mechanism in embryonic development are limited. This study assessed the toxicity of sodium fluoride (NaF) toward zebrafish embryos. We determined the mortality, hatching rate, phenotypic malformation, heart function, and morphology of zebrafish embryos after exposure to NaF. Subsequently, the molecular mechanism was revealed using high-throughput RNA sequencing analysis. The expression levels of key genes for heart development were detected using quantitative real-time reverse transcription PCR. The 50% lethal concentration (LC50) value of NaF toward zebrafish embryos at 96 h post-fertilization was 335.75 mg/L. When the concentration of NaF was higher than 200 mg/L, severe deformities, such as pericardial edema, yolk sac edema, spine curvature, shortened body length, reduced head area, and eye area, were observed. The heart rate of the embryos exposed to NaF decreased in a dose-dependent fashion. The distance between the sinus venosus and bulbus arteriosus was significantly increased in the NaF-exposed group compared with that in the control group. The stroke volume and cardiac output decreased significantly in the NaF groups. Compared with the control group, the expression levels of Gata4, Tbx5a, Hand2, Tnnt2c, Nppa, and Myh6 were significantly increased in the NaF-treated group. Through transcriptome sequencing, 1354 differentially expressed genes (DEGs) were detected in the NaF (200 mg/L) treated groups, including 1253 upregulated genes and 101 downregulated genes. Gene ontology functional analysis and Kyoto Encyclopedia of Genes and Genomes pathway analyses of the DEGs showed that cardiac-related pathways, such as actin cytoskeleton regulation, Jak-Stat, PI3k-Akt, and Ras, were activated in the NaF-exposed group. This study revealed the underlying mechanism of fluoride-induced cardiac morphological and functional abnormalities and provides clues for the clinical prevention and treatment of fluorosis.
Collapse
Affiliation(s)
- Feiqing Wang
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin City, 300072, China
| | - Fa Chen
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Wen Song
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Yanju Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Haiyan Wu
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Tingting Tian
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Mengxian Tian
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Dongxin Tang
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China.
| | - Yang Liu
- Clinical Research Center, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Bao Shan North Road, Yunyan District, Guiyang, 550001, Guizhou Province, China.
| |
Collapse
|
8
|
Riabkova NS, Kogan AE, Katrukha IA, Vylegzhanina AV, Bogomolova AP, Alieva AK, Pevzner DV, Bereznikova AV, Katrukha AG. Influence of Anticoagulants on the Dissociation of Cardiac Troponin Complex in Blood Samples. Int J Mol Sci 2024; 25:8919. [PMID: 39201603 PMCID: PMC11354535 DOI: 10.3390/ijms25168919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Immunodetection of cardiac isoforms of troponin I (cTnI) and troponin T (cTnT) in blood samples is widely used for the diagnosis of acute myocardial infarction. The cardiac troponin complex (ITC-complex), comprising cTnI, cTnT, and troponin C (TnC), makes up a large portion of troponins released into the bloodstream after the necrosis of cardiomyocytes. However, the stability of the ITC-complex has not been fully investigated. This study aimed to investigate the stability of the ITC-complex in blood samples. A native ITC-complex was incubated in buffer solutions, serum, and citrate, heparin, or EDTA plasma at various temperatures. Western blotting and gel filtration were performed, and troponins were detected using specific monoclonal antibodies. The ITC-complex dissociated at 37 °C in buffers with or without anticoagulants, in citrate, heparin, and EDTA plasmas, and in serum, into a binary cTnI-TnC complex (IC-complex) and free cTnT. In plasma containing heparin and EDTA, the IC-complex further dissociated into free TnC and cTnI. No dissociation was found at 4 °C or at room temperature (RT) in all matrices within 24 h except for EDTA plasma. After incubation at 37 °C in EDTA plasma and serum, dissociation was accompanied by proteolytic degradation of both cTnI and cTnT. The presence of anti-troponin autoantibodies in the sample impeded dissociation of the ITC-complex. The ITC-complex dissociates in vitro to form the IC-complex and free cTnT at 37 °C but is mostly stable at 4 °C or RT. Further dissociation of the IC-complex occurs at 37 °C in plasmas containing heparin and EDTA.
Collapse
Affiliation(s)
- Natalia S. Riabkova
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
- Department of Biochemistry, Faculty of Biology, Moscow State University, Leninskie Gory 1, str. 12, 119234 Moscow, Russia
| | - Alexander E. Kogan
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
- Department of Biochemistry, Faculty of Biology, Moscow State University, Leninskie Gory 1, str. 12, 119234 Moscow, Russia
| | - Ivan A. Katrukha
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
- Department of Biochemistry, Faculty of Biology, Moscow State University, Leninskie Gory 1, str. 12, 119234 Moscow, Russia
| | - Alexandra V. Vylegzhanina
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
| | - Agnessa P. Bogomolova
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
- Department of Biochemistry, Faculty of Biology, Moscow State University, Leninskie Gory 1, str. 12, 119234 Moscow, Russia
| | - Amina K. Alieva
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Akademik Chazov str., 15A, 121552 Moscow, Russia; (A.K.A.); (D.V.P.)
| | - Dmitry V. Pevzner
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Akademik Chazov str., 15A, 121552 Moscow, Russia; (A.K.A.); (D.V.P.)
| | - Anastasia V. Bereznikova
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
- Department of Biochemistry, Faculty of Biology, Moscow State University, Leninskie Gory 1, str. 12, 119234 Moscow, Russia
| | - Alexey G. Katrukha
- HyTest Ltd., Intelligate 1, 6th Floor, Joukahaisenkatu 6, 20520 Turku, Finland; (N.S.R.); (A.E.K.); (A.V.V.); (A.P.B.); (A.V.B.); (A.G.K.)
- Department of Biochemistry, Faculty of Biology, Moscow State University, Leninskie Gory 1, str. 12, 119234 Moscow, Russia
| |
Collapse
|
9
|
Kula AJ, Albers E, Hong B, Kemna M, Friedland-Little J, Law Y. Trajectories of postoperative serum troponin concentrations following pediatric heart transplantation. JHLT OPEN 2024; 5:100039. [PMID: 40143913 PMCID: PMC11935518 DOI: 10.1016/j.jhlto.2023.100039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 03/28/2025]
Abstract
Background Troponin is a biomarker of myocardial injury and death but has not been well studied after pediatric heart transplants. The objective of this analysis is to describe the distribution and clinical determinants of serum troponin measured in the first week after pediatric heart transplantation. Methods We included all patients who underwent heart transplantation at Seattle Children's Hospital between 2012 and 2016. Serum Troponin-I (TnI) was measured daily in the first week after transplant. We described the distribution of serum TnI, and examined the relationship between peak TnI with known pre- peri-operative risk factors for myocardial injury including etiology of heart failure, ischemia time, and donor to recipient characteristics. Logistic regression models were used to test the association between peak TnI with incidence of death or rejection and formation of donor-specific antibodies (DSA) within 1 year. Adjusted models included age, HF etiology, crossmatch status, and panel reactive antibodies. Results During the study period, 86 transplants were performed on 83 unique individuals. Serum TnI peaked at a median of 0.9 days after transplantation. In adjusted models, higher peak TnI was associated with death and/or rejection within 1-year post-transplant (odds ratio [95% confidence interval]: 1.10 [1.02, 1.19]). Peak TnI was not associated with de-novo DSA formation in adjusted models (OR [95%CI]: 1.01 [0.94, 1.09]). Post-transplant length of stay in the intensive care unit was positively correlated with peak TnI (r = 0.36, p < 0.001). Conclusions This study describes serum TnI in the first week after pediatric heart transplant; a population for whom existing data are sparse. Our findings suggest TnI may have utility as a readily measurable biomarker of transplant-related myocardial injury. These results may inform future investigations of the prognostic significance of higher post-transplant TnI in future studies.
Collapse
Affiliation(s)
- Alexander J. Kula
- Division of Nephrology, Ann & Robert H. Lurie Children’s Hospital of Chicago, and Department of Pediatrics, Northwestern University, Chicago, Illinois
- Division of Cardiology, Seattle Children's Hospital, and Department of Pediatrics, University of Washington, Seattle, Washington
| | - Erin Albers
- Department of Biostatistics, University of Washington, Seattle, Washington
- Division of Cardiology, Seattle Children's Hospital, and Department of Pediatrics, University of Washington, Seattle, Washington
| | - Bora Hong
- Department of Biostatistics, University of Washington, Seattle, Washington
- Division of Cardiology, Seattle Children's Hospital, and Department of Pediatrics, University of Washington, Seattle, Washington
| | - Mariska Kemna
- Department of Biostatistics, University of Washington, Seattle, Washington
- Division of Cardiology, Seattle Children's Hospital, and Department of Pediatrics, University of Washington, Seattle, Washington
| | - Joshua Friedland-Little
- Department of Biostatistics, University of Washington, Seattle, Washington
- Division of Cardiology, Seattle Children's Hospital, and Department of Pediatrics, University of Washington, Seattle, Washington
| | - Yuk Law
- Department of Biostatistics, University of Washington, Seattle, Washington
- Division of Cardiology, Seattle Children's Hospital, and Department of Pediatrics, University of Washington, Seattle, Washington
| |
Collapse
|
10
|
Celeski M, Segreti A, Piscione M, Monticelli LM, Di Gioia G, Fossati C, Ussia GP, Pigozzi F, Grigioni F. The current paradigm of cardiac troponin increase among athletes. Monaldi Arch Chest Dis 2024. [PMID: 38700130 DOI: 10.4081/monaldi.2024.2878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/16/2024] [Indexed: 05/05/2024] Open
Abstract
Although it is known that exercise improves cardiovascular health and extends life expectancy, a significant number of people may also experience an elevation in cardiac troponin levels as a result of exercise. For many years, researchers have argued whether exercise-induced cardiac troponin rises are a consequence of a physiological or pathological reaction and whether they are clinically significant. Differences in cardiac troponin elevation and cardiac remodeling can be seen between athletes participating in different types of sports. When forecasting the exercise-induced cardiac troponin rise, there are many additional parameters to consider, as there is a large amount of interindividual heterogeneity in the degree of cardiac troponin elevation. Although it was previously believed that cardiac troponin increases in athletes represented a benign phenomenon, numerous recent studies disproved this notion by demonstrating that, in specific individuals, cardiac troponin increases may have clinical and prognostic repercussions. This review aims to examine the role of cardiac troponin in athletes and its role in various sporting contexts. This review also discusses potential prognostic and clinical implications, as well as future research methods, and provides a straightforward step-by-step algorithm to help clinicians interpret cardiac troponin rise in athletes in both ischemic and non-ischemic circumstances.
Collapse
Affiliation(s)
- Mihail Celeski
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome.
| | - Andrea Segreti
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome; Department of Movement, Human and Health Sciences, University of Rome "Foro Italico".
| | - Mariagrazia Piscione
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome.
| | - Luigi Maria Monticelli
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome.
| | - Giuseppe Di Gioia
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome; Department of Movement, Human and Health Sciences, University of Rome "Foro Italico"; Institute of Sport Medicine and Science, Italian National Olympic Committee, Rome.
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico".
| | - Gian Paolo Ussia
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome.
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico".
| | - Francesco Grigioni
- Research Unit of Cardiovascular Science, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma; Cardiology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome.
| |
Collapse
|
11
|
Newman JD, Anthopolos R, Ruggles KV, Cornwell M, Reynolds HR, Bangalore S, Mavromatis K, Held C, Wallentin L, Kullo IJ, McManus B, Newby LKK, Rosenberg Y, Hochman JS, Maron DJ, Berger JS. Biomarkers and cardiovascular events in patients with stable coronary disease in the ISCHEMIA Trials. Am Heart J 2023; 266:61-73. [PMID: 37604357 PMCID: PMC10843480 DOI: 10.1016/j.ahj.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/23/2023]
Abstract
IMPORTANCE Biomarkers may improve prediction of cardiovascular events for patients with stable coronary artery disease (CAD), but their importance in addition to clinical tests of inducible ischemia and CAD severity is unknown. OBJECTIVES To evaluate the prognostic value of multiple biomarkers in stable outpatients with obstructive CAD and moderate or severe inducible ischemia. DESIGN AND SETTING The ISCHEMIA and ISCHEMIA CKD trials randomized 5,956 participants with CAD to invasive or conservative management from July 2012 to January 2018; 1,064 participated in the biorepository. MAIN OUTCOME MEASURES Primary outcome was cardiovascular death, myocardial infarction (MI), or hospitalization for unstable angina, heart failure, or resuscitated cardiac arrest. Secondary outcome was cardiovascular death or MI. Improvements in prediction were assessed by cause-specific hazard ratios (HR) and area under the receiver operating characteristics curve (AUC) for an interquartile increase in each biomarker, controlling for other biomarkers, in a base clinical model of risk factors, left ventricular ejection fraction (LVEF) and ischemia severity. Secondary analyses were performed among patients in whom core-lab confirmed severity of CAD was ascertained by computed cardiac tomographic angiography (CCTA). EXPOSURES Baseline levels of interleukin-6 (IL-6), high sensitivity troponin T (hsTnT), growth differentiation factor 15 (GDF-15), N-terminal pro-B-type natriuretic peptide (NT-proBNP), lipoprotein a (Lp[a]), high sensitivity C-reactive protein (hsCRP), Cystatin C, soluble CD 40 ligand (sCD40L), myeloperoxidase (MPO), and matrix metalloproteinase 3 (MMP3). RESULTS Among 757 biorepository participants, median (IQR) follow-up was 3 (2-5) years, age was 67 (61-72) years, and 144 (19%) were female; 508 had severity of CAD by CCTA available. In an adjusted multimarker model with hsTnT, GDF-15, NT-proBNP and sCD40L, the adjusted HR for the primary outcome per interquartile increase in each biomarker was 1.58 (95% CI 1.22, 2.205), 1.60 (95% CI 1.16, 2.20), 1.61 (95% 1.22, 2.14), and 1.46 (95% 1.12, 1.90), respectively. The adjusted multimarker model also improved prediction compared with the clinical model, increasing the AUC from 0.710 to 0.792 (P < .01) and 0.714 to 0.783 (P < .01) for the primary and secondary outcomes, respectively. Similar findings were observed after adjusting for core-lab confirmed atherosclerosis severity. CONCLUSIONS AND RELEVANCE Among ISCHEMIA biorepository participants, biomarkers of myocyte injury/distension, inflammation, and platelet activity improved cardiovascular event prediction in addition to risk factors, LVEF, and assessments of ischemia and atherosclerosis severity. These biomarkers may improve risk stratification for patients with stable CAD.
Collapse
Affiliation(s)
- Jonathan D Newman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY.
| | - Rebecca Anthopolos
- Division of Biostatistics, Department of Population Health, NYU Langone Health, New York, NY
| | - Kelly V Ruggles
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | | | | | - Sripal Bangalore
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Kreton Mavromatis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Iftikar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Bruce McManus
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Kristin K Newby
- Division of Cardiology, Department of Medicine, Duke Clinical Research Institute, Durham, NC
| | - Yves Rosenberg
- Division of Cardiovascular Sciences, National Health Lung and Blood Institute, National Institute of Health, Bethesda, MD
| | - Judith S Hochman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - David J Maron
- Department of Medicine, Stanford University, Stanford, CA
| | - Jeffrey S Berger
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
12
|
Peppa M, Manta A, Mavroeidi I, Asimakopoulou A, Syrigos A, Nastos C, Pikoulis E, Kollias A. Changes in Cardiovascular and Renal Biomarkers Associated with SGLT2 Inhibitors Treatment in Patients with Type 2 Diabetes Mellitus. Pharmaceutics 2023; 15:2526. [PMID: 38004506 PMCID: PMC10675228 DOI: 10.3390/pharmaceutics15112526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/26/2023] Open
Abstract
Type 2 diabetes mellitus is a major health problem worldwide with a steadily increasing prevalence reaching epidemic proportions. The major concern is the increased morbidity and mortality due to diabetic complications. Traditional but also nontraditional risk factors have been proposed to explain the pathogenesis of type 2 diabetes mellitus and its complications. Hyperglycemia has been considered an important risk factor, and the strict glycemic control can have a positive impact on microangiopathy but not macroangiopathy and its related morbidity and mortality. Thus, the therapeutic algorithm has shifted focus from a glucose-centered approach to a strategy that now emphasizes target-organ protection. Sodium-glucose transporter 2 inhibitors is an extremely important class of antidiabetic medications that, in addition to their glucose lowering effect, also exhibit cardio- and renoprotective effects. Various established and novel biomarkers have been described, reflecting kidney and cardiovascular function. In this review, we investigated the changes in established but also novel biomarkers of kidney, heart and vascular function associated with sodium-glucose transporter 2 inhibitors treatment in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Melpomeni Peppa
- Endocrine Unit, 2nd Propaedeutic Department of Internal Medicine, School of Medicine, Research Institute and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, 12641 Athens, Greece; (A.M.); (I.M.)
- 3rd Department of Internal Medicine, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.A.); (A.S.); (A.K.)
| | - Aspasia Manta
- Endocrine Unit, 2nd Propaedeutic Department of Internal Medicine, School of Medicine, Research Institute and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, 12641 Athens, Greece; (A.M.); (I.M.)
| | - Ioanna Mavroeidi
- Endocrine Unit, 2nd Propaedeutic Department of Internal Medicine, School of Medicine, Research Institute and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, 12641 Athens, Greece; (A.M.); (I.M.)
| | - Athina Asimakopoulou
- 3rd Department of Internal Medicine, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.A.); (A.S.); (A.K.)
| | - Alexandros Syrigos
- 3rd Department of Internal Medicine, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.A.); (A.S.); (A.K.)
| | - Constantinos Nastos
- 3rd Department of Surgery, School of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12641 Athens, Greece; (C.N.); (E.P.)
| | - Emmanouil Pikoulis
- 3rd Department of Surgery, School of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12641 Athens, Greece; (C.N.); (E.P.)
| | - Anastasios Kollias
- 3rd Department of Internal Medicine, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.A.); (A.S.); (A.K.)
| |
Collapse
|
13
|
El-Sayed SAES, El-Alfy ES, Sayed-Ahmed MZ, Mohanta UK, Alqahtani SS, Alam N, Ahmad S, Ali MS, Igarashi I, Rizk MA. Evaluating the inhibitory effect of resveratrol on the multiplication of several Babesia species and Theileria equi on in vitro cultures, and Babesia microti in mice. Front Pharmacol 2023; 14:1192999. [PMID: 37324476 PMCID: PMC10267976 DOI: 10.3389/fphar.2023.1192999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Histone post-translational modification is one of the most studied factors influencing epigenetic regulation of protozoan parasite gene expression, which is mediated by histone deacetylases (KDACs) and acetyltransferases (KATs). Objective and methods: The present study investigated the role of resveratrol (RVT) as an activator of histone deacetylases in the control of various pathogenic Babesia sp. and Theileria equi in vitro, as well as B. microti infected mice in vivo using fluorescence assay. Its role in mitigating the side effects associated with the widely used antibabesial drugs diminazene aceturate (DA) and azithromycin (AZM) has also been investigated. Results: The in vitro growth of B. bovis, B. bigemina, B. divergens, B. caballi and Theileria equi (T. equi) was significantly inhibited (P < 0.05) by RVT treatments. The estimated IC50 values revealed that RVT has the greatest inhibitory effects on B. bovis growth in vitro, with an IC50 value of 29.51 ± 2.46 µM. Reverse transcription PCR assay showed that such inhibitory activity might be attributed to resveratrol's stimulatory effect on B. bovis KDAC3 (BbKADC3) as well as its inhibitory effect on BbKATS. RVT causes a significant decrease (P < 0.05) in cardiac troponin T (cTnT) levels in heart tissue of B. microti- infected mice, thereby indicating that RVT may play a part in reducing the cardiotoxic effects of AZM. Resveratrol showed an additive effect with imidocarb dipropionate in vivo. Treatment of B. microti-infected mice with a combined 5 mg/kg RVT and 8.5 mg/kg ID resulted in an 81.55% inhibition at day 10 postinoculation (peak of parasitemia). Conclusion: Our data show that RVT is a promising antibabesial pharmacological candidate with therapeutic activities that could overcome the side effects of the currently used anti-Babesia medications.
Collapse
Affiliation(s)
- Shimaa Abd El-Salam El-Sayed
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - El-Sayed El-Alfy
- Parasitology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Z. Sayed-Ahmed
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jizan, Saudi Arabia
| | - Uday Kumar Mohanta
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Saad S. Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Nawazish Alam
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jizan, Saudi Arabia
| | - Sarfaraz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jizan, Saudi Arabia
| | - Md Sajid Ali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jizan, Saudi Arabia
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Mohamed Abdo Rizk
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
14
|
Joolharzadeh P, Rodriguez M, Zaghlol R, Pedersen LN, Jimenez J, Bergom C, Mitchell JD. Recent Advances in Serum Biomarkers for Risk Stratification and Patient Management in Cardio-Oncology. Curr Cardiol Rep 2023; 25:133-146. [PMID: 36790618 PMCID: PMC9930715 DOI: 10.1007/s11886-022-01834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 02/16/2023]
Abstract
PURPOSE OF REVIEW Following significant advancements in cancer therapeutics and survival, the risk of cancer therapy-related cardiotoxicity (CTRC) is increasingly recognized. With ongoing efforts to reduce cardiovascular morbidity and mortality in cancer patients and survivors, cardiac biomarkers have been studied for both risk stratification and monitoring during and after therapy to detect subclinical disease. This article will review the utility for biomarker use throughout the cancer care continuum. RECENT FINDINGS A recent meta-analysis shows utility for troponin in monitoring patients at risk for CTRC during cancer therapy. The role for natriuretic peptides is less clear but may be useful in patients receiving proteasome inhibitors. Early studies explore use of myeloperoxidase, growth differentiation factor 15, galectin 3, micro-RNA, and others as novel biomarkers in CTRC. Biomarkers have potential to identify subclinical CTRC and may reveal opportunities for early intervention. Further research is needed to elucidate optimal biomarkers and surveillance strategies.
Collapse
Affiliation(s)
- Pouya Joolharzadeh
- General Medical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Mario Rodriguez
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO, USA
| | - Raja Zaghlol
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO, USA
| | - Lauren N Pedersen
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jesus Jimenez
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Bergom
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Joshua D Mitchell
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO, USA.
- Alvin J. Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
15
|
Abrams ST, Alhamdi Y, Zi M, Guo F, Du M, Wang G, Cartwright EJ, Toh CH. Extracellular Histone-Induced Protein Kinase C Alpha Activation and Troponin Phosphorylation Is a Potential Mechanism of Cardiac Contractility Depression in Sepsis. Int J Mol Sci 2023; 24:ijms24043225. [PMID: 36834636 PMCID: PMC9967552 DOI: 10.3390/ijms24043225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Reduction in cardiac contractility is common in severe sepsis. However, the pathological mechanism is still not fully understood. Recently it has been found that circulating histones released after extensive immune cell death play important roles in multiple organ injury and disfunction, particularly in cardiomyocyte injury and contractility reduction. How extracellular histones cause cardiac contractility depression is still not fully clear. In this work, using cultured cardiomyocytes and a histone infusion mouse model, we demonstrate that clinically relevant histone concentrations cause significant increases in intracellular calcium concentrations with subsequent activation and enriched localization of calcium-dependent protein kinase C (PKC) α and βII into the myofilament fraction of cardiomyocytes in vitro and in vivo. Furthermore, histones induced dose-dependent phosphorylation of cardiac troponin I (cTnI) at the PKC-regulated phosphorylation residues (S43 and T144) in cultured cardiomyocytes, which was also confirmed in murine cardiomyocytes following intravenous histone injection. Specific inhibitors against PKCα and PKCβII revealed that histone-induced cTnI phosphorylation was mainly mediated by PKCα activation, but not PKCβII. Blocking PKCα also significantly abrogated histone-induced deterioration in peak shortening, duration and the velocity of shortening, and re-lengthening of cardiomyocyte contractility. These in vitro and in vivo findings collectively indicate a potential mechanism of histone-induced cardiomyocyte dysfunction driven by PKCα activation with subsequent enhanced phosphorylation of cTnI. These findings also indicate a potential mechanism of clinical cardiac dysfunction in sepsis and other critical illnesses with high levels of circulating histones, which holds the potential translational benefit to these patients by targeting circulating histones and downstream pathways.
Collapse
Affiliation(s)
- Simon T. Abrams
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
| | - Yasir Alhamdi
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield S5 7AU, UK
| | - Min Zi
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Fengmei Guo
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- The Medical School, Southeast University, Nanjing 210009, China
| | - Min Du
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
| | - Guozheng Wang
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| | - Elizabeth J. Cartwright
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Roald Dahl Haemostasis & Thrombosis Centre, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| |
Collapse
|
16
|
Risi CM, Belknap B, White HD, Dryden K, Pinto JR, Chase PB, Galkin VE. High-resolution cryo-EM structure of the junction region of the native cardiac thin filament in relaxed state. PNAS NEXUS 2023; 2:pgac298. [PMID: 36712934 PMCID: PMC9832952 DOI: 10.1093/pnasnexus/pgac298] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Cardiac contraction depends on molecular interactions among sarcomeric proteins coordinated by the rising and falling intracellular Ca2+ levels. Cardiac thin filament (cTF) consists of two strands composed of actin, tropomyosin (Tm), and equally spaced troponin (Tn) complexes forming regulatory units. Tn binds Ca2+ to move Tm strand away from myosin-binding sites on actin to enable actomyosin cross-bridges required for force generation. The Tn complex has three subunits-Ca2+-binding TnC, inhibitory TnI, and Tm-binding TnT. Tm strand is comprised of adjacent Tm molecules that overlap "head-to-tail" along the actin filament. The N-terminus of TnT (e.g., TnT1) binds to the Tm overlap region to form the cTF junction region-the region that connects adjacent regulatory units and confers to cTF internal cooperativity. Numerous studies have predicted interactions among actin, Tm, and TnT1 within the junction region, although a direct structural description of the cTF junction region awaited completion. Here, we report a 3.8 Å resolution cryo-EM structure of the native cTF junction region at relaxing (pCa 8) Ca2+ conditions. We provide novel insights into the "head-to-tail" interactions between adjacent Tm molecules and interactions between the Tm junction with F-actin. We demonstrate how TnT1 stabilizes the Tm overlap region via its interactions with the Tm C- and N-termini and actin. Our data show that TnT1 works as a joint that anchors the Tm overlap region to actin, which stabilizes the relaxed state of the cTF. Our structure provides insight into the molecular basis of cardiac diseases caused by missense mutations in TnT1.
Collapse
Affiliation(s)
- Cristina M Risi
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Kelly Dryden
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22904, USA
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
17
|
Leite L, Matos P, Leon-Justel A, Espírito-Santo C, Rodríguez-Padial L, Rodrigues F, Orozco D, Redon J. High sensitivity troponins: A potential biomarkers of cardiovascular risk for primary prevention. Front Cardiovasc Med 2022; 9:1054959. [PMID: 36531726 PMCID: PMC9748104 DOI: 10.3389/fcvm.2022.1054959] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/14/2022] [Indexed: 03/07/2024] Open
Abstract
There have been several approaches to building charts for CV risk, all of which have both strengths and limitations. Identifying early organ damage provides relevant information and should be included in risk charts, although the direct relationship with risk is imprecise, variability between operators at the time to assess, and low availability in some healthcare systems, limits its use. Biomarkers, like troponin (cTns) isoforms cTnI and cTnT, a cardiac specific myocyte injury marker, have the great advantage of being relatively reproducible, more readily accessible, and applicable to different populations. New and improved troponin assays have good analytical performance, can measure very low levels of circulating troponin, and have low intra individual variation, below 10 %. Several studies have analyzed the blood levels in healthy subjects and their predictive value for cardiovascular events in observational, prospective and post-hoc studies. All of them offered relevant information and shown that high sensitivity hs-cTnI has a place as an additional clinical marker to add to current charts, and it also reflects sex- and age-dependent differences. Although few more questions need to be answered before recommend cTnI for assessing CV risk in primary prevention, seems to be a potential strong marker to complement CV risk charts.
Collapse
Affiliation(s)
- Luis Leite
- Cardiology Department, Coimbra University Hospital, University of Coimbra, Coimbra, Portugal
| | - Pedro Matos
- APDP e Hospital CUF Infante Santo, Lisbon, Portugal
| | - Antonio Leon-Justel
- Department of Laboratory Medicine, Virgen Macarena University Hospital, Seville, Spain
| | | | | | | | - Domingo Orozco
- Department of Clinical Medicine, Miguel Hernández University, Elche, Spain
| | - Josep Redon
- INCLIVA Research Institute, University of Valencia, Valencia, Spain
- CIBERObn Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Ji K, Jiao D, Yang G, Degen AA, Zhou J, Liu H, Wang W, Cong H. Transcriptome analysis revealed potential genes involved in thermogenesis in muscle tissue in cold-exposed lambs. Front Genet 2022; 13:1017458. [PMID: 36338953 PMCID: PMC9634817 DOI: 10.3389/fgene.2022.1017458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
Cold tolerance is an important trait for sheep raised at high altitudes. Muscle tissue, comprising 30–40% of the total body mass, produces heat during cold exposure. However, little is known about the genetic mechanisms of this tissue and its role in thermogenesis in lambs. We examined genes in skeletal muscle tissue in a cold-adapted sheep breed, Altay, and a cold-intolerant sheep breed, Hu, when exposed to low air temperature. Three ewe-lambs of each breed were maintained at −5°C and three ewe-lambs of each breed were maintained at 20°C. After cold exposure for 25 days, the longissimus dorsi of each lamb was collected, and transcriptome profiles were sequenced and analyzed. The results of RNA-seq showed that the average reads among the four groups were 11.0 Gbase. The genome mapping rate averaged 88.1% and the gene mapping rate averaged 82.5%. The analysis of differentially expressed genes (DEGs) indicated that the peroxisome proliferator-activated receptors (PPAR), cAMP, and calcium signaling pathways and muscle contraction in muscle tissue were linked to thermogenesis in cold-exposed lambs. Furthermore, PCK1 (phosphoenolpyruvate carboxykinase1) increased glyceroneogenesis in cold-exposed Altay lambs, and APOC3 (apolipoprotein C3), LPL (lipoprotein lipase), and FABP4 (fatty acid binding protein 4, adipocyte) were involved in the intake and transport of free fatty acids. In Hu sheep, cAMP biosynthesis from ATP hydrolysis was regulated by ADCY10 (adenylate cyclase) and ADORA2a (adenosine A2a receptor). Skeletal muscle contraction was regulated by MYL2 (myosin light chain 2). In conclusion, cold exposure altered the expression level of genes involved in heat production in muscle tissue. Some potential mechanisms were revealed, including calcium ion transport in the calcium signaling pathway, fatty acid metabolism in the PPAR signaling pathway, and cAMP biosynthesis in the cAMP signaling pathway. This study implied that skeletal muscle plays an important role in thermoregulation in lambs.
Collapse
Affiliation(s)
- Kaixi Ji
- Key Laboratory of Stress Physiology and Ecology of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dan Jiao
- Key Laboratory of Stress Physiology and Ecology of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China
| | - Guo Yang
- Key Laboratory of Stress Physiology and Ecology of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, China
- *Correspondence: Guo Yang,
| | - Abraham Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of Negev, Beer Sheva, Israel
| | - Jianwei Zhou
- State Key Laboratory of Grassland and Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Hu Liu
- College of Ecology, Lanzhou University, Lanzhou, China
| | - Wenqiang Wang
- College of Ecology, Lanzhou University, Lanzhou, China
| | - Haitao Cong
- Dongying Modern Animal Husbandry Development Service Center, Dongying, China
| |
Collapse
|
19
|
Kawana M, Spudich JA, Ruppel KM. Hypertrophic cardiomyopathy: Mutations to mechanisms to therapies. Front Physiol 2022; 13:975076. [PMID: 36225299 PMCID: PMC9548533 DOI: 10.3389/fphys.2022.975076] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/22/2022] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) affects more than 1 in 500 people in the general population with an extensive burden of morbidity in the form of arrhythmia, heart failure, and sudden death. More than 25 years since the discovery of the genetic underpinnings of HCM, the field has unveiled significant insights into the primary effects of these genetic mutations, especially for the myosin heavy chain gene, which is one of the most commonly mutated genes. Our group has studied the molecular effects of HCM mutations on human β-cardiac myosin heavy chain using state-of-the-art biochemical and biophysical tools for the past 10 years, combining insights from clinical genetics and structural analyses of cardiac myosin. The overarching hypothesis is that HCM-causing mutations in sarcomere proteins cause hypercontractility at the sarcomere level, and we have shown that an increase in the number of myosin molecules available for interaction with actin is a primary driver. Recently, two pharmaceutical companies have developed small molecule inhibitors of human cardiac myosin to counteract the molecular consequences of HCM pathogenesis. One of these inhibitors (mavacamten) has recently been approved by the FDA after completing a successful phase III trial in HCM patients, and the other (aficamten) is currently being evaluated in a phase III trial. Myosin inhibitors will be the first class of medication used to treat HCM that has both robust clinical trial evidence of efficacy and that targets the fundamental mechanism of HCM pathogenesis. The success of myosin inhibitors in HCM opens the door to finding other new drugs that target the sarcomere directly, as we learn more about the genetics and fundamental mechanisms of this disease.
Collapse
Affiliation(s)
- Masataka Kawana
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States,*Correspondence: Kathleen M. Ruppel,
| |
Collapse
|
20
|
Bay B, Goßling A, Blaum CM, Kroeger F, Koppe L, Lorenz T, Koester L, Clemmensen P, Westermann D, Kirchhof P, Blankenberg S, Zeller T, Seiffert M, Waldeyer C, Brunner FJ. Association of High-Sensitivity Troponin T and I Blood Concentrations With All-Cause Mortality and Cardiovascular Outcome in Stable Patients-Results From the INTERCATH Cohort. J Am Heart Assoc 2022; 11:e024516. [PMID: 35862141 PMCID: PMC9496405 DOI: 10.1161/jaha.121.024516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 06/21/2022] [Indexed: 02/03/2023]
Abstract
Background The association between high-sensitivity troponin T (hsTnT) and high-sensitivity troponin I (hsTnI) and outcome when adjusted for confounders including the angiographical severity of coronary artery disease (CAD) remains largely unknown. We therefore aimed to explore whether hsTnT and hsTnI blood levels increase with CAD severity and add independent predictive information for future major adverse cardiovascular events and all-cause mortality in stable patients. Methods and Results Patients from the INTERCATH cohort with available coronary angiography and hsTnT and hsTnI concentrations were included. Troponin concentrations were quantified via hsTnT (Roche Elecsys) and hsTnI (Abbott ARCHITECT STAT). To investigate the association of hsTnT and hsTnI with outcome, a multivariable analysis adjusting for classical cardiovascular risk factors, low-density lipoprotein cholesterol, estimated glomerular filtration rate, hs-CRP (high-sensitivity C-reactive protein), NT-proBNP (N-terminal pro-brain natriuretic peptide), and Gensini score was carried out. Of 1829 patients, 27.9% were women, and the mean age was 68.6±10.9 years. Troponin blood concentrations were higher in patients with diagnosed CAD compared with those without. Using a linear regression model current smoking, arterial hypertension, estimated glomerular filtration rate, hs-CRP, NT-proBNP, and CAD severity as graded by the Gensini and SYNTAX scores were associated with high-sensitivity troponin levels. Patients were followed for 4.4 years (25th and 75th percentiles: 4.3, 4.4). After multivariable adjustment, all-cause mortality was predicted by hsTnT (hazard ratio [HR], 1.7 [95% CI, 1.5-2.2], P<0.001) as well as hsTnI (HR, 1.5 [95% CI, 1.2-1.8], P<0.001). However, only hsTnI (HR, 1.2 [95% CI, 1.0-1.4], P=0.032) remained as an independent predictor of major adverse cardiovascular events after adjusting for most possible confounders, including CAD severity (hsTnT: HR, 1.0 [95% CI, 0.9-1.2], P=0.95). Conclusions After adjusting for classical cardiovascular risk factors, low-density lipoprotein cholesterol, estimated glomerular filtration rate, hs-CRP, NT-proBNP, and CAD severity, hsTnT and hsTnI were independently associated with all-cause mortality, but only hsTnI was associated with major adverse cardiovascular events in stable patients undergoing coronary angiography. Registration URL: https://clinicaltrials.gov/; Unique identifier: NCT04936438.
Collapse
Affiliation(s)
- Benjamin Bay
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Alina Goßling
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christopher M. Blaum
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Friederike Kroeger
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Luise Koppe
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Thiess Lorenz
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Lukas Koester
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Peter Clemmensen
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
- Department of Regional Health Research, Faculty of Health SciencesUniversity of Southern Denmark and Nykoebing Falster HospitalOdenseDenmark
| | - Dirk Westermann
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Paulus Kirchhof
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Stefan Blankenberg
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Tanja Zeller
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Moritz Seiffert
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Christoph Waldeyer
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Fabian J. Brunner
- Department of CardiologyUniversity Heart & Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckHamburgGermany
| |
Collapse
|
21
|
Cyclic Stretching Induces Maturation of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes through Nuclear-Mechanotransduction. Tissue Eng Regen Med 2022; 19:781-792. [PMID: 35258794 PMCID: PMC9294081 DOI: 10.1007/s13770-021-00427-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 11/22/2021] [Accepted: 12/21/2021] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND During cardiogenesis, cardiac cells receive various stimuli, such as biomechanical and chemical cues, from the surrounding microenvironment, and these signals induce the maturation of heart cells. Mechanical force, especially tensile force in the heart, is one of the key stimuli that induce cardiomyocyte (CM) maturation through mechanotransduction, a process through which physical cues are transformed into biological responses. However, the effects and mechanisms of tensile force on cell maturation are poorly studied. METHODS In this study, we developed a cyclic stretch system that mimics the mechanical environment of the heart by loading tensile force to human-induced pluripotent stem cell (hiPSC)-derived CMs. hiPSC-CMs cultured with the cyclic stretch system analyzed morphological change, immunofluorescent staining, expression of maturation markers in mRNA, and beating properties compared to static cultures. RESULTS hiPSC-CMs cultured with the cyclic stretch system showed increased cell alignment, sarcomere length and expression of maturation markers in mRNA, such as TNNI3, MYL2 and TTN, compared to static cultures. Especially, the expression of genes related to nuclear mechanotransduction, such as Yap1, Lamin A/C, plectin, and desmin, was increased in the cyclically stretched hiPSC-CMs. Furthermore, the volume of the nucleus was increased by as much as 120% in the cyclic stretch group. CONCLUSION These results revealed that nuclear mechanotransduction induced by tensile force is involved in CM maturation. Together, these findings provide novel evidence suggesting that nuclear mechanotransduction induced by tensile force is involved in the regulation of cardiac maturation.
Collapse
|
22
|
Senekovič Kojc T, Marčun Varda N. Novel Biomarkers of Heart Failure in Pediatrics. CHILDREN 2022; 9:children9050740. [PMID: 35626917 PMCID: PMC9139970 DOI: 10.3390/children9050740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023]
Abstract
Novel biomarkers of heart failure are the subject of numerous studies. Biomarkers of heart failure can be determined in the blood and in the urine. Seven groups of biomarkers of heart failure based on pathophysiological mechanisms are presented in this review, namely biomarkers of myocardial stretch, myocyte injury, myocardial remodeling, biomarkers of inflammation, renal dysfunction, neurohumoral activation, and oxidative stress. Studies of biomarkers in the pediatric population are scarce, therefore, further investigation is needed for reliable prognostic and therapeutic implications. The future of biomarker use is in multimarker panels that include a combination of biomarkers with different pathophysiological mechanisms in order to improve their diagnostic and prognostic predictive value.
Collapse
Affiliation(s)
- Teja Senekovič Kojc
- Department of Perinatology, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia
- Correspondence:
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, Taborska 8, 2000 Maribor, Slovenia
| |
Collapse
|
23
|
Hulikova A, Park KC, Loonat AA, Gunadasa-Rohling M, Curtis MK, Chung YJ, Wilson A, Carr CA, Trafford AW, Fournier M, Moshnikova A, Andreev OA, Reshetnyak YK, Riley PR, Smart N, Milne TA, Crump NT, Swietach P. Alkaline nucleoplasm facilitates contractile gene expression in the mammalian heart. Basic Res Cardiol 2022; 117:17. [PMID: 35357563 PMCID: PMC8971196 DOI: 10.1007/s00395-022-00924-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 01/31/2023]
Abstract
Cardiac contractile strength is recognised as being highly pH-sensitive, but less is known about the influence of pH on cardiac gene expression, which may become relevant in response to changes in myocardial metabolism or vascularization during development or disease. We sought evidence for pH-responsive cardiac genes, and a physiological context for this form of transcriptional regulation. pHLIP, a peptide-based reporter of acidity, revealed a non-uniform pH landscape in early-postnatal myocardium, dissipating in later life. pH-responsive differentially expressed genes (pH-DEGs) were identified by transcriptomics of neonatal cardiomyocytes cultured over a range of pH. Enrichment analysis indicated "striated muscle contraction" as a pH-responsive biological process. Label-free proteomics verified fifty-four pH-responsive gene-products, including contractile elements and the adaptor protein CRIP2. Using transcriptional assays, acidity was found to reduce p300/CBP acetylase activity and, its a functional readout, inhibit myocardin, a co-activator of cardiac gene expression. In cultured myocytes, acid-inhibition of p300/CBP reduced H3K27 acetylation, as demonstrated by chromatin immunoprecipitation. H3K27ac levels were more strongly reduced at promoters of acid-downregulated DEGs, implicating an epigenetic mechanism of pH-sensitive gene expression. By tandem cytoplasmic/nuclear pH imaging, the cardiac nucleus was found to exercise a degree of control over its pH through Na+/H+ exchangers at the nuclear envelope. Thus, we describe how extracellular pH signals gain access to the nucleus and regulate the expression of a subset of cardiac genes, notably those coding for contractile proteins and CRIP2. Acting as a proxy of a well-perfused myocardium, alkaline conditions are permissive for expressing genes related to the contractile apparatus.
Collapse
Affiliation(s)
- Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Kyung Chan Park
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Aminah A Loonat
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - M Kate Curtis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Yu Jin Chung
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Abigail Wilson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Marjorie Fournier
- Department of Biochemistry, Advanced Proteomics Facility, University of Oxford, Oxford, UK
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Oleg A Andreev
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Yana K Reshetnyak
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Thomas A Milne
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford, UK
| | - Nicholas T Crump
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
24
|
Ronen D, Bsoul A, Lotem M, Abedat S, Yarkoni M, Amir O, Asleh R. Exploring the Mechanisms Underlying the Cardiotoxic Effects of Immune Checkpoint Inhibitor Therapies. Vaccines (Basel) 2022; 10:vaccines10040540. [PMID: 35455289 PMCID: PMC9031363 DOI: 10.3390/vaccines10040540] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Adaptive immune response modulation has taken a central position in cancer therapy in recent decades. Treatment with immune checkpoint inhibitors (ICIs) is now indicated in many cancer types with exceptional results. The two major inhibitory pathways involved are cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and programmed cell death protein 1 (PD-1). Unfortunately, immune activation is not tumor-specific, and as a result, most patients will experience some form of adverse reaction. Most immune-related adverse events (IRAEs) involve the skin and gastrointestinal (GI) tract; however, any organ can be involved. Cardiotoxicity ranges from arrhythmias to life-threatening myocarditis with very high mortality rates. To date, most treatments of ICI cardiotoxicity include immune suppression, which is also not cardiac-specific and may result in hampering of tumor clearance. Understanding the mechanisms behind immune activation in the heart is crucial for the development of specific treatments. Histological data and other models have shown mainly CD4 and CD8 infiltration during ICI-induced cardiotoxicity. Inhibition of CTLA4 seems to result in the proliferation of more diverse T0cell populations, some of which with autoantigen recognition. Inhibition of PD-1 interaction with PD ligand 1/2 (PD-L1/PD-L2) results in release from inhibition of exhausted self-recognizing T cells. However, CTLA4, PD-1, and their ligands are expressed on a wide range of cells, indicating a much more intricate mechanism. This is further complicated by the identification of multiple co-stimulatory and co-inhibitory signals, as well as the association of myocarditis with antibody-driven myasthenia gravis and myositis IRAEs. In this review, we focus on the recent advances in unraveling the complexity of the mechanisms driving ICI cardiotoxicity and discuss novel therapeutic strategies for directly targeting specific underlying mechanisms to reduce IRAEs and improve outcomes.
Collapse
Affiliation(s)
- Daniel Ronen
- Department of Internal Medicine D, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Aseel Bsoul
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
| | - Michal Lotem
- Department of Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
| | - Merav Yarkoni
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
- Correspondence: ; Tel.: +972-2-6776564; Fax: +972-2-6411028
| |
Collapse
|
25
|
Reinwald H, Alvincz J, Salinas G, Schäfers C, Hollert H, Eilebrecht S. Toxicogenomic profiling after sublethal exposure to nerve- and muscle-targeting insecticides reveals cardiac and neuronal developmental effects in zebrafish embryos. CHEMOSPHERE 2022; 291:132746. [PMID: 34748799 DOI: 10.1016/j.chemosphere.2021.132746] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/15/2021] [Accepted: 10/29/2021] [Indexed: 06/13/2023]
Abstract
For specific primary modes of action (MoA) in environmental non-target organisms, EU legislation restricts the usage of active substances of pesticides or biocides. Corresponding regulatory hazard assessments are costly, time consuming and require large numbers of non-human animal studies. Currently, predictive toxicology of development compounds relies on their chemical structure and provides little insights into toxicity mechanisms that precede adverse effects. Using the zebrafish embryo model, we characterized transcriptomic responses to a range of sublethal concentrations of six nerve- and muscle-targeting insecticides with different MoA (abamectin, carbaryl, chlorpyrifos, fipronil, imidacloprid & methoxychlor). Our aim was to identify affected biological processes and suitable biomarker candidates for MoA-specific signatures. Abamectin showed the most divergent signature among the tested insecticides, linked to lipid metabolic processes. Differentially expressed genes (DEGs) after imidacloprid exposure were primarily associated with immune system and inflammation. In total, 222 early responsive genes to either MoA were identified, many related to three major processes: (1) cardiac muscle cell development and functioning (tcap, desma, bag3, hspb1, hspb8, flnca, myoz3a, mybpc2b, actc2, tnnt2c), (2) oxygen transport and hypoxic stress (alas2, hbbe1.1, hbbe1.3, hbbe2, hbae3, igfbp1a, hif1al) and (3) neuronal development and plasticity (npas4a, egr1, btg2, ier2a, vgf). The thyroidal function related gene dio3b was upregulated by chlorpyrifos and downregulated by higher abamectin concentrations. Important regulatory genes for cardiac muscle (tcap) and forebrain development (npas4a) were the most frequently ifferentially expressed across all insecticide treatments. We consider the identified gene sets as useful early warning biomarker candidates, i.e. for developmental toxicity targeting heart and brain in aquatic vertebrates. Our findings provide a better understanding about early molecular events in response to the analyzed MoA. Perceptively, this promotes the development for sensitive and informative biomarker-based in vitro assays for toxicological MoA prediction and AOP refinement, without the suffering of adult fish.
Collapse
Affiliation(s)
- Hannes Reinwald
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany; Department Evolutionary Ecology and Environmental Toxicology, Faculty Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Julia Alvincz
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Gabriela Salinas
- NGS-Services for Integrative Genomics, University of Göttingen, Göttingen, Germany
| | - Christoph Schäfers
- Department of Ecotoxicology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Henner Hollert
- Department Evolutionary Ecology and Environmental Toxicology, Faculty Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Sebastian Eilebrecht
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany.
| |
Collapse
|
26
|
Siddiq MM, Chan AT, Miorin L, Yadaw AS, Beaumont KG, Kehrer T, Cupic A, White KM, Tolentino RE, Hu B, Stern AD, Tavassoly I, Hansen J, Sebra R, Martinez P, Prabha S, Dubois N, Schaniel C, Iyengar-Kapuganti R, Kukar N, Giustino G, Sud K, Nirenberg S, Kovatch P, Albrecht RA, Goldfarb J, Croft L, McLaughlin MA, Argulian E, Lerakis S, Narula J, García-Sastre A, Iyengar R. Functional Effects of Cardiomyocyte Injury in COVID-19. J Virol 2022; 96:e0106321. [PMID: 34669512 PMCID: PMC8791272 DOI: 10.1128/jvi.01063-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/18/2021] [Indexed: 01/19/2023] Open
Abstract
COVID-19 affects multiple organs. Clinical data from the Mount Sinai Health System show that substantial numbers of COVID-19 patients without prior heart disease develop cardiac dysfunction. How COVID-19 patients develop cardiac disease is not known. We integrated cell biological and physiological analyses of human cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the presence of interleukins (ILs) with clinical findings related to laboratory values in COVID-19 patients to identify plausible mechanisms of cardiac disease in COVID-19 patients. We infected hiPSC-derived cardiomyocytes from healthy human subjects with SARS-CoV-2 in the absence and presence of IL-6 and IL-1β. Infection resulted in increased numbers of multinucleated cells. Interleukin treatment and infection resulted in disorganization of myofibrils, extracellular release of troponin I, and reduced and erratic beating. Infection resulted in decreased expression of mRNA encoding key proteins of the cardiomyocyte contractile apparatus. Although interleukins did not increase the extent of infection, they increased the contractile dysfunction associated with viral infection of cardiomyocytes, resulting in cessation of beating. Clinical data from hospitalized patients from the Mount Sinai Health System show that a significant portion of COVID-19 patients without history of heart disease have elevated troponin and interleukin levels. A substantial subset of these patients showed reduced left ventricular function by echocardiography. Our laboratory observations, combined with the clinical data, indicate that direct effects on cardiomyocytes by interleukins and SARS-CoV-2 infection might underlie heart disease in COVID-19 patients. IMPORTANCE SARS-CoV-2 infects multiple organs, including the heart. Analyses of hospitalized patients show that a substantial number without prior indication of heart disease or comorbidities show significant injury to heart tissue, assessed by increased levels of troponin in blood. We studied the cell biological and physiological effects of virus infection of healthy human iPSC-derived cardiomyocytes in culture. Virus infection with interleukins disorganizes myofibrils, increases cell size and the numbers of multinucleated cells, and suppresses the expression of proteins of the contractile apparatus. Viral infection of cardiomyocytes in culture triggers release of troponin similar to elevation in levels of COVID-19 patients with heart disease. Viral infection in the presence of interleukins slows down and desynchronizes the beating of cardiomyocytes in culture. The cell-level physiological changes are similar to decreases in left ventricular ejection seen in imaging of patients' hearts. These observations suggest that direct injury to heart tissue by virus can be one underlying cause of heart disease in COVID-19.
Collapse
Affiliation(s)
- Mustafa M. Siddiq
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Angel T. Chan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Arjun S. Yadaw
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas Kehrer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kris M. White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rosa E. Tolentino
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bin Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alan D. Stern
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Iman Tavassoly
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jens Hansen
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Sema4, a Mount Sinai Venture, Stamford, Connecticut, USA
| | - Pedro Martinez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Som Prabha
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicole Dubois
- Department of Cell Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christoph Schaniel
- Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Hematology & Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rupa Iyengar-Kapuganti
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nina Kukar
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gennaro Giustino
- Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Hematology & Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Karan Sud
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sharon Nirenberg
- Department of Scientific Computing and Data Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patricia Kovatch
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Scientific Computing and Data Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Randy A. Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joseph Goldfarb
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lori Croft
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maryann A. McLaughlin
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Edgar Argulian
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stamatios Lerakis
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jagat Narula
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
27
|
Chaulin AM. Some Common Causes of False Positive Increases in Serum Levels of Cardiac Troponins. Curr Cardiol Rev 2022; 18:e130422203527. [PMID: 35422225 PMCID: PMC9893154 DOI: 10.2174/1573403x18666220413124038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/21/2022] [Accepted: 02/26/2022] [Indexed: 11/22/2022] Open
Abstract
Cardiac troponin molecules (cTnI and cTnT) are the most valuable and in-demand biomarkers for detecting various types of myocardial damage (reversible and irreversible, ischemic, inflammatory, toxic, etc.) in current clinical practice. These biomarkers are widely used for early diagnosis of acute myocardial infarction (AMI) and risk stratification of patients suffering from a number of cardiac (such as myocarditis, heart failure, cardiomyopathy, etc.) and extra-cardiac diseases (such as sepsis, renal failure, pulmonary embolism, neurological pathologies, etc.) that negatively affect the cells of cardiac muscle tissue. However, in daily routine clinical activities, internists and cardiologists often encounter cases of false increases in the concentrations of cardiospecific troponins. A false increase in the concentration of troponins contributes to an incorrect diagnosis and incorrect therapy, which can harm the patient. A false increase in the concentration of troponins contributes to an incorrect diagnosis and incorrect therapy, which can harm the patient, therefore, internists and cardiologists should be well aware of the main reasons and mechanisms for false-positive results cTnI and cTnT. This review article mainly focuses on the causes of falsepositive increases in serum levels of cTnI and cTnT, which provide helpful clues for the accurate diagnosis of AMI and evidence for the differential diagnosis.
Collapse
Affiliation(s)
- Aleksey Michailovich Chaulin
- Department of Cardiology and Cardiovascular Surgery, Medical Faculty, Samara State Medical University, Samara, Russia
- Department of Clinical Chemistry, Samara Regional Clinical Cardiological Dispensary, Samara, Russia
| |
Collapse
|
28
|
Xu T, Meng QH, Gilchrist SC, Lin SH, Lin R, Xu T, Milgrom SA, Gandhi SJ, Wu H, Zhao Y, Lopez-Mattei JC, Mohan R, Liao Z. Assessment of Prognostic Value of High-Sensitivity Cardiac Troponin T for Early Prediction of Chemoradiation Therapy-Induced Cardiotoxicity in Patients with Non-Small Cell Lung Cancer: A Secondary Analysis of a Prospective Randomized Trial. Int J Radiat Oncol Biol Phys 2021; 111:907-916. [PMID: 34302893 PMCID: PMC8530972 DOI: 10.1016/j.ijrobp.2021.07.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/14/2021] [Accepted: 07/12/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE Cardiotoxicities induced by cancer therapy can negatively affect quality of life and survival. We investigated whether high-sensitivity cardiac troponin T (hs-cTnT) levels could serve as biomarker for early detection of cardiac adverse events (CAEs) after chemoradiation therapy (CRT) for non-small cell lung cancer (NSCLC). METHODS AND MATERIALS This study included 225 patients who received concurrent platinum and taxane-doublet chemotherapy with thoracic radiation therapy to a total dose of 60 to 74 Gy for NSCLC. All patients were evaluated for CAEs; 190 patients also had serial hs-cTnT measurements. RESULTS Grade ≥3 CAEs occurred in 24 patients (11%) at a median interval of 9 months after CRT. Pretreatment hs-cTnT levels were higher in men, in patients aged ≥64 years, and in patients with pre-existing heart disease or poor performance status (P < .05). hs-cTnT levels increased at 4 weeks during CRT (P < .05) and decreased after completion of CRT but did not return to pretreatment levels (P = .002). The change (Δ) in hs-cTnT levels during CRT correlated with mean heart dose (P = .0004), the heart volumes receiving 5 to 55 Gy (P < .05), and tumor location (P = .006). Risks of severe CAEs and mortality were significantly increased if the pretreatment hs-cTnT was >10 ng/L or the Δ during CRT was ≥5 ng/L. CONCLUSIONS Elevation of hs-cTnT during CRT was radiation heart dose-dependent, and high hs-cTnT levels during the course of CRT were associated with CAEs and mortality. Routine monitoring of hs-cTnT could identify patients who are at high risk of CRT-induced CAEs early to guide modifications of cancer therapy and possible interventions to mitigate cardiotoxicity.
Collapse
Affiliation(s)
- Ting Xu
- Departments of Radiation Oncology
| | | | | | | | - Ruitao Lin
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tianlin Xu
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sarah A Milgrom
- Department of Radiation Oncology, University of Colorado-Denver, Denver, Colorado
| | | | - Haijun Wu
- Department of Radiation Oncology, Xiangya Hospital, Xiangya Medical School of Central South University, Hunan, P. R. of China
| | - Yu Zhao
- Department of Medicine, University Hospital, Rochester, New York
| | | | - Radhe Mohan
- Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
29
|
Hassoun R, Budde H, Mügge A, Hamdani N. Cardiomyocyte Dysfunction in Inherited Cardiomyopathies. Int J Mol Sci 2021; 22:11154. [PMID: 34681814 PMCID: PMC8541428 DOI: 10.3390/ijms222011154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023] Open
Abstract
Inherited cardiomyopathies form a heterogenous group of disorders that affect the structure and function of the heart. Defects in the genes encoding sarcomeric proteins are associated with various perturbations that induce contractile dysfunction and promote disease development. In this review we aimed to outline the functional consequences of the major inherited cardiomyopathies in terms of myocardial contraction and kinetics, and to highlight the structural and functional alterations in some sarcomeric variants that have been demonstrated to be involved in the pathogenesis of the inherited cardiomyopathies. A particular focus was made on mutation-induced alterations in cardiomyocyte mechanics. Since no disease-specific treatments for familial cardiomyopathies exist, several novel agents have been developed to modulate sarcomere contractility. Understanding the molecular basis of the disease opens new avenues for the development of new therapies. Furthermore, the earlier the awareness of the genetic defect, the better the clinical prognostication would be for patients and the better the prevention of development of the disease.
Collapse
Affiliation(s)
- Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
30
|
Arangalage D, Degrauwe N, Michielin O, Monney P, Özdemir BC. Pathophysiology, diagnosis and management of cardiac toxicity induced by immune checkpoint inhibitors and BRAF and MEK inhibitors. Cancer Treat Rev 2021; 100:102282. [PMID: 34438238 DOI: 10.1016/j.ctrv.2021.102282] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/16/2022]
Abstract
Immune checkpoint inhibitors (ICIs) and BRAF and MEK inhibitors (BRAFi/MEKi) have drastically improved the outcome of melanoma patients. ICIs can induce myocarditis, a rare immune related adverse event (irAE) with an estimated lethality of 50%. BRAFi/MEKi may induce left ventricular ejection fraction decrease, hypertension or QT interval prolongation. While the BRAFi/MEKi induced cardiotoxicity is often reversible upon treatment discontinuation or dose adaptation and symptomatic therapy is often sufficient to restore cardiac function, the treatment of ICI-induced myocarditis mainly relies on high dose corticosteroids. There is no established therapy for steroid resistant myocarditis, yet various drugs have been reported to improve outcome. Shared epitopes between melanoma cells and cardiac tissue are thought to underlie the development of ICIs induced myocarditis. The mechanism of BRAFi/MEKi induced cardiotoxicity appears to be related to the Ras-Raf-MEK-ERK pathway in cardiomyocyte repair, survival and proliferation. With the emerging application of ICI-BRAFi/MEKi combinations, so called triplet therapies, differentiating between these two types of cardiotoxicity will become important for appropriate patient management. In this article we provide a summary of the existing literature on the pathophysiology, diagnosis and management of cardiotoxicity of melanoma therapies.
Collapse
Affiliation(s)
- Dimitri Arangalage
- Department of Cardiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Nils Degrauwe
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Pierre Monney
- Department of Cardiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Berna C Özdemir
- Department of Oncology, Bern University Hospital (Inselspital), University of Bern, Switzerland; International Cancer Prevention Institute, Epalinges, Switzerland.
| |
Collapse
|
31
|
Inconsistent Findings of Cardiac Troponin T and I in Clinical Routine Diagnostics: Factors of Influence. J Clin Med 2021; 10:jcm10143148. [PMID: 34300313 PMCID: PMC8305654 DOI: 10.3390/jcm10143148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/05/2021] [Accepted: 07/14/2021] [Indexed: 11/17/2022] Open
Abstract
Cardiac troponins are crucial for the diagnosis of acute myocardial infarction. Despite known differences in their diagnostic implication, there are no recommendations for only one of the two troponins, cardiac troponin I (cTnI) and troponin T (cTnT) so far. In an everyday routine diagnostic, cTnT (Roche) as well as cTnI (Abbott) were measured in 5667 samples from 3264 patient cases. We investigated the number of identical or discrepant troponin findings. Regarding cTnI, we considered both, sex-dependent and unisex cutoffs. In particular, the number of cTnT positive and cTnI negative results was strikingly high in 14.0% of cTnT positive samples and increases to 23.8% by using sex-specific cTnI cutoffs. This group was considerably greater than the group of cTnI positive and cTnT negative results, also after elimination of patients with an eGFR < 60 mL/min/1.73 m2. Comparing the troponin cases with a dynamic increase or decrease between two measurements, we saw a balanced number of discrepant cases (between cTnT+/cTnI- and cTnT-/cTnI+), which was, however, still present. Using ROC analysis, sex-dependent cutoffs improved sensitivity and specificity of cTnI. This study shows in a large cohort that comparing the two cardiac troponins does not amount to identical analytical results. Consideration of sex-dependent cutoffs may improve sensitivity and specificity.
Collapse
|
32
|
Taranova NA, Slobodenuyk VD, Zherdev AV, Dzantiev BB. Network of gold conjugates for enhanced sensitive immunochromatographic assays of troponins. RSC Adv 2021; 11:16445-16452. [PMID: 35479181 PMCID: PMC9030257 DOI: 10.1039/d1ra02212a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/27/2021] [Indexed: 01/13/2023] Open
Abstract
Highly sensitive detection of cardiac troponins I and T (cTnI and cTnT) was completed by immunochromatography with double amplification, through the binding of functionalized gold nanoparticles (GNPs). The robust nature of the approach, based on the formation of nanoparticle networks through the biotin-streptavidin interaction, was confirmed; the choice of the best assay parameters for maximal increase in ICA sensitivity was demonstrated. A bifunctional conjugate of GNPs with biotinylated specific IgG and two auxiliary conjugates, GNP-biotin and GNP-streptavidin, form three-component aggregates in the analytical zone of the test strip. The inclusion of abundant gold labels in the resulting immune complex leads to an amplified colorimetric signal. The limits of detection (LoDs) of cTnI and cTnT were 0.9 and 0.4 ng mL-1, respectively, which is 3 times lower than the LoDs of more commonly used systems. Visual LoDs were 10-fold lower in concentration. The enhancement has been realized both in single and double assay formats; analysis of cTnI and cTnT presented the same characteristics.
Collapse
Affiliation(s)
- Nadezhda A Taranova
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences Leninsky Prospect 33 119071 Moscow Russia
| | - Vladislav D Slobodenuyk
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences Leninsky Prospect 33 119071 Moscow Russia
| | - Anatoly V Zherdev
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences Leninsky Prospect 33 119071 Moscow Russia
| | - Boris B Dzantiev
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences Leninsky Prospect 33 119071 Moscow Russia
| |
Collapse
|
33
|
Rayani K, Seffernick J, Li AY, Davis JP, Spuches AM, Van Petegem F, Solaro RJ, Lindert S, Tibbits GF. Binding of calcium and magnesium to human cardiac troponin C. J Biol Chem 2021; 296:100350. [PMID: 33548225 PMCID: PMC7961095 DOI: 10.1016/j.jbc.2021.100350] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 01/03/2023] Open
Abstract
Cardiac muscle thin filaments are composed of actin, tropomyosin, and troponin that change conformation in response to Ca2+ binding, triggering muscle contraction. Human cardiac troponin C (cTnC) is the Ca2+-sensing component of the thin filament. It contains structural sites (III/IV) that bind both Ca2+ and Mg2+ and a regulatory site (II) that has been thought to bind only Ca2+. Binding of Ca2+ at this site initiates a series of conformational changes that culminate in force production. However, the mechanisms that underpin the regulation of binding at site II remain unclear. Here, we have quantified the interaction between site II and Ca2+/Mg2+ through isothermal titration calorimetry and thermodynamic integration simulations. Direct and competitive binding titrations with WT N-terminal cTnC and full-length cTnC indicate that physiologically relevant concentrations of both Ca2+/Mg2+ interacted with the same locus. Moreover, the D67A/D73A N-terminal cTnC construct in which two coordinating residues within site II were removed was found to have significantly reduced affinity for both cations. In addition, 1 mM Mg2+ caused a 1.4-fold lower affinity for Ca2+. These experiments strongly suggest that cytosolic-free Mg2+ occupies a significant population of the available site II. Interaction of Mg2+ with site II of cTnC likely has important functional consequences for the heart both at baseline as well as in diseased states that decrease or increase the availability of Mg2+, such as secondary hyperparathyroidism or ischemia, respectively.
Collapse
Affiliation(s)
- Kaveh Rayani
- Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Justin Seffernick
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, Ohio, USA
| | - Alison Yueh Li
- Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| | - Anne Marie Spuches
- Department of Chemistry, East Carolina University, 300 Science and Technology Building, Greenville, North Carolina, USA
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - R John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, Ohio, USA
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Simon Fraser University, Burnaby, British Columbia, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada; Cardiac Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.
| |
Collapse
|
34
|
Han CR, Wang H, Hoffmann V, Zerfas P, Kruhlak M, Cheng SY. Thyroid Hormone Receptor α Mutations Cause Heart Defects in Zebrafish. Thyroid 2021; 31:315-326. [PMID: 32762296 PMCID: PMC7891307 DOI: 10.1089/thy.2020.0332] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Mutations of thyroid hormone receptor α1 (TRα1) cause resistance to thyroid hormone (RTHα). Patients exhibit growth retardation, delayed bone development, anemia, and bradycardia. By using mouse models of RTHα, much has been learned about the molecular actions of TRα1 mutants that underlie these abnormalities in adults. Using zebrafish models of RTHα that we have recently created, we aimed to understand how TRα1 mutants affect the heart function during this period. Methods: In contrast to human and mice, the thra gene is duplicated, thraa and thrab, in zebrafish. Using CRISPR/Cas9-mediated targeted mutagenesis, we created C-terminal mutations in each of two duplicated thra genes in zebrafish (thraa 8-bp insertion or thrab 1-bp insertion mutations). We recently showed that these mutant fish faithfully recapitulated growth retardation as found in patients and thra mutant mice. In the present study, we used histological analysis, gene expression profiles, confocal fluorescence, and transmission electron microscopy (TEM) to comprehensively analyze the phenotypic characteristics of mutant fish heart during development. Results: We found both a dilated atrium and an abnormally shaped ventricle in adult mutant fish. The retention of red blood cells in the two abnormal heart chambers, and the decreased circulating blood speed and reduced expression of contractile genes indicated weakened contractility in the heart of mutant fish. These abnormalities were detected in mutant fish as early as 35 days postfertilization (juveniles). Furthermore, the expression of genes associated with the sarcomere assembly was suppressed in the heart of mutant fish, resulting in abnormalities of sarcomere organization as revealed by TEM, suggesting that the abnormal sarcomere organization could underlie the bradycardia exhibited in mutant fish. Conclusions: Using a zebrafish model of RTHα, the present study demonstrated for the first time that TRα1 mutants could act to cause abnormal heart structure, weaken contractility, and disrupt sarcomere organization that affect heart functions. These findings provide new insights into the bradycardia found in RTHα patients.
Collapse
Affiliation(s)
- Cho Rong Han
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hui Wang
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Victoria Hoffmann
- Diagnostic and Research Services Branch, Office of Research Services, National Institutes of Health, Bethesda, Maryland, USA
| | - Patricia Zerfas
- Diagnostic and Research Services Branch, Office of Research Services, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Kruhlak
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Address correspondence to: Sheue-Yann Cheng, PhD, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5128, Bethesda, MD 20892-4264, USA
| |
Collapse
|
35
|
Song Y, Cai X, Ostermeyer G, Yu J, Du D, Lin Y. Self-Assembling Allochroic Nanocatalyst for Improving Nanozyme-Based Immunochromatographic Assays. ACS Sens 2021; 6:220-228. [PMID: 33433202 DOI: 10.1021/acssensors.0c02148] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Paper-based rapid diagnostic tests, such as immunochromatographic assays, namely lateral flow immunoassay (LFA), are valuable alternatives for biomarker detection compared to traditional laboratory-based tests, but these assays need further refinement to consolidate their biosensing capabilities. Nanozyme integration into LFA systems may provide a reliable means of improving the analytic sensitivity of LFA tests. Due to the involvement of multiple liquid-handling steps, the quantitative accuracy is compromised, hence hindering the use of untrained personnel point-of-care use. Self-assembling allochroic nanocatalyst (SAN) assemblies satisfy these LFA quality measures by optimizing analyte-antibody reporting performance and by intrinsically catalyzing chromogen activation, thereby reducing the number of liquid handling steps involved during sample analysis. In SANs, the hydrophobic chromogens serve as peroxidase substrates that self-assemble into nanoparticles at high loading fractions. These features demonstrate the potential for SAN-LFAs to be a valuable patient point-of-care (POC) test. Herein, we describe the SAN fabrication process and employ SAN-LFAs to detect cardiac troponin I-troponin C (cTnI-TnC) and myoglobin (Myo) levels present in plasma samples. Using SAN-LFAs, the limits of detection for cTnI-TnC and Myo were 0.012 ng/mL and 0.2 ng/mL respectively. We also demonstrate SAN compatibility with blood samples and stability under long-term storage conditions. The successful utlization of SANs in LFA-based biomarker detection may inspire these nanocatalysts to be integrated into similar immunochromatographic testing methods.
Collapse
Affiliation(s)
- Yang Song
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99164, United States
- Immuno Diagnostics Division and Biomonitoring, Evergreen System Inc., Seattle, Washington 98105, United States
| | - Xiaoli Cai
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99164, United States
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry Central China Normal University, Wuhan 430079, PR China
| | - Grayson Ostermeyer
- School of Biological Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Jierui Yu
- Department of Chemistry and Biochemistry, Southern Illinois University, Carbondale, Illinois 62901, United States
| | - Dan Du
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Yuehe Lin
- School of Mechanical and Material Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
36
|
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: 40] [Impact Index Per Article: 8.0] [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
|
37
|
Siddiq MM, Chan AT, Miorin L, Yadaw AS, Beaumont KG, Kehrer T, White KM, Cupic A, Tolentino RE, Hu B, Stern AD, Tavassoly I, Hansen J, Martinez P, Dubois N, Schaniel C, Iyengar-Kapuganti R, Kukar N, Giustino G, Sud K, Nirenberg S, Kovatch P, Goldfarb J, Croft L, McLaughlin MA, Argulian E, Lerakis S, Narula J, García-Sastre A, Iyengar R. Physiology of cardiomyocyte injury in COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.10.20229294. [PMID: 33200140 PMCID: PMC7668750 DOI: 10.1101/2020.11.10.20229294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
COVID-19 affects multiple organs. Clinical data from the Mount Sinai Health System shows that substantial numbers of COVID-19 patients without prior heart disease develop cardiac dysfunction. How COVID-19 patients develop cardiac disease is not known. We integrate cell biological and physiological analyses of human cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) infected with SARS-CoV-2 in the presence of interleukins, with clinical findings, to investigate plausible mechanisms of cardiac disease in COVID-19 patients. We infected hiPSC-derived cardiomyocytes, from healthy human subjects, with SARS-CoV-2 in the absence and presence of interleukins. We find that interleukin treatment and infection results in disorganization of myofibrils, extracellular release of troponin-I, and reduced and erratic beating. Although interleukins do not increase the extent, they increase the severity of viral infection of cardiomyocytes resulting in cessation of beating. Clinical data from hospitalized patients from the Mount Sinai Health system show that a significant portion of COVID-19 patients without prior history of heart disease, have elevated troponin and interleukin levels. A substantial subset of these patients showed reduced left ventricular function by echocardiography. Our laboratory observations, combined with the clinical data, indicate that direct effects on cardiomyocytes by interleukins and SARS-CoV-2 infection can underlie the heart disease in COVID-19 patients.
Collapse
Affiliation(s)
- Mustafa M. Siddiq
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Angel T. Chan
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
- Departments of Medicine and Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lisa Miorin
- Department of Microbiology and Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Arjun S. Yadaw
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Thomas Kehrer
- Department of Microbiology and Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Kris M. White
- Department of Microbiology and Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Anastasija Cupic
- Department of Microbiology and Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Rosa E. Tolentino
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Bin Hu
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Alan D. Stern
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Iman Tavassoly
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Jens Hansen
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Pedro Martinez
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Nicole Dubois
- Department of Cell Developmental and Regenerative Biology and Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Christoph Schaniel
- Division of Hematology & Oncology Department of Medicine and Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Rupa Iyengar-Kapuganti
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Nina Kukar
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Gennaro Giustino
- Division of Hematology & Oncology Department of Medicine and Black Family Stem Cell Center, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Karan Sud
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Sharon Nirenberg
- Department of Scientific Computing and Data Science, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Patricia Kovatch
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY 10029
- Department of Scientific Computing and Data Science, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Joseph Goldfarb
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Lori Croft
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Maryann A. McLaughlin
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Edgar Argulian
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Stamatios Lerakis
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Jagat Narula
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Adolfo García-Sastre
- Department of Microbiology and Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York NY 10029
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029
| | - Ravi Iyengar
- Department of Pharmacological Sciences, and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York NY 10029
| |
Collapse
|
38
|
Liu L, Zhang D, Li Y. LncRNAs in cardiac hypertrophy: From basic science to clinical application. J Cell Mol Med 2020; 24:11638-11645. [PMID: 32896990 PMCID: PMC7579708 DOI: 10.1111/jcmm.15819] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a typical pathological phenotype of cardiomyopathy and a result from pathological remodelling of cardiomyocytes in humans. At present, emerging evidence demonstrated the roles of long non‐coding RNAs (lncRNAs) in regulating the pathophysiological process of cardiac hypertrophy. Herein, we would like to review the recent researches on this issue and try to analysis the potential therapeutic targets on lncRNA sites. Studies have revealed both genetic mutations related hypertrophic cardiomyopathy and the compensative cardiac hypertrophy due to pressure overload, inflammation, endocrine issues and other external stimulations, share a common molecular mechanism of ventricular hypertrophy. The emerging evidence identified the abnormal expression of lncRNAs would leading to the impairment the function of sarcomere, intracellular calcium handling and mitochondrial metabolisms. Several researches proved the therapeutic role of lncRNAs in preventing or reversing cardiac hypertrophy. With the development of delivery system for small pieces of oligonucleotide, clinicians could design gene therapy approaches to terminate the process of cardiac hypertrophy to provide better prognosis.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Characterization and validation of a preventative therapy for hypertrophic cardiomyopathy in a murine model of the disease. Proc Natl Acad Sci U S A 2020; 117:23113-23124. [PMID: 32859761 PMCID: PMC7502707 DOI: 10.1073/pnas.2002976117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hypertrophic cardiomyopathy affects 1:500 of the general population. Current drug therapy is used to manage symptoms in patients. There is an unmet need for treatments that can prevent the cardiomyopathy. Here we identify biomarkers of hypertrophic cardiomyopathy resulting from causing cardiac troponin I mutation Gly203Ser, and present a safe, nontoxic, preventative approach for the treatment of associated cardiomyopathy. Currently there is an unmet need for treatments that can prevent hypertrophic cardiomyopathy (HCM). Using a murine model we previously identified that HCM causing cardiac troponin I mutation Gly203Ser (cTnI-G203S) is associated with increased mitochondrial metabolic activity, consistent with the human condition. These alterations precede development of the cardiomyopathy. Here we examine the efficacy of in vivo treatment of cTnI-G203S mice with a peptide derived against the α-interaction domain of the cardiac L-type calcium channel (AID-TAT) on restoring mitochondrial metabolic activity, and preventing HCM. cTnI-G203S or age-matched wt mice were treated with active or inactive AID-TAT. Following treatment, targeted metabolomics was utilized to evaluate myocardial substrate metabolism. Cardiac myocyte mitochondrial metabolic activity was assessed as alterations in mitochondrial membrane potential and flavoprotein oxidation. Cardiac morphology and function were examined using echocardiography. Cardiac uptake was assessed using an in vivo multispectral imaging system. We identified alterations in six biochemical intermediates in cTnI-G203S hearts consistent with increased anaplerosis. We also reveal that AID-TAT treatment of precardiomyopathic cTnI-G203S mice, but not mice with established cardiomyopathy, restored cardiac myocyte mitochondrial membrane potential and flavoprotein oxidation, and prevented myocardial hypertrophy. Importantly, AID-TAT was rapidly targeted to the heart, and not retained by the liver or kidneys. Overall, we identify biomarkers of HCM resulting from the cTnI mutation Gly203Ser, and present a safe, preventative therapy for associated cardiomyopathy. Utilizing AID-TAT to modulate cardiac metabolic activity may be beneficial in preventing HCM in “at risk” patients with identified Gly203Ser gene mutations.
Collapse
|
40
|
Hosseinifard H, Ghadimi N, Kaveh S, Shabaninejad H, Lijassi A, Azarfarin R. Comparing cardiac troponin levels using sevoflurane and isoflurane in patients undergoing cardiac surgery: a systematic review and meta-analysis. J Cardiovasc Thorac Res 2020; 12:1-9. [PMID: 32211131 PMCID: PMC7080340 DOI: 10.34172/jcvtr.2020.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 01/10/2020] [Indexed: 01/02/2023] Open
Abstract
Introduction: Cardiac troponin is one of the heart biomarkers and its high levels correlates with a high risk of cardiomyocytes damage. This study aimed to compare sevoflurane and isoflurane effect on troponin levels in patients undergoing cardiac surgery. Methods: We systematically searched for RCTs which had been published in Cochrane library, PubMed, Web of science, CRD, Scopus, and Google Scholar by the end of February 30th, 2019. The quality of articles was evaluated with the Cochrane checklist. GRADE was used for quality of evidence for this meta-analysis. Meta-analysis was done based on random or fixed effect model. Results: Five studies with total of 190 (sevoflurane) and 191 (isoflurane) patients were included. The results showed that pooled mean difference of troponin levels between the two groups was significant at ICU admission time and 24 hours after entering. The comparison of troponin level changes between the two groups (baseline = at time ICU) in 24 and 48 hours after ICU admission was significant. Conclusion: This meta-analysis showed that blood troponin levels were significantly lower at the time of arrival in ICU with isoflurane and after 24 hours with sevoflurane. Generally, given the small mean difference between isoflurane and sevoflurane, it seems that none of the medications has a negative effect on the cardiac troponin level.
Collapse
Affiliation(s)
- Hossein Hosseinifard
- Biostatistics, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Nashmil Ghadimi
- Health Technology Assessment, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Kaveh
- Health Technology Assessment, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Shabaninejad
- Department of Health Services Management, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran.,Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Alaadine Lijassi
- Faculty of Medicine and Pharmacy of Rabat, Mohammed V University of Rabat, Rabat, Morocco
| | - Rasoul Azarfarin
- Echocardiography Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Fletcher S, Maddock H, James RS, Wallis R, Gharanei M. The cardiac work-loop technique: An in vitro model for identifying and profiling drug-induced changes in inotropy using rat papillary muscles. Sci Rep 2020; 10:5258. [PMID: 32210283 PMCID: PMC7093439 DOI: 10.1038/s41598-020-58935-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/26/2019] [Indexed: 11/09/2022] Open
Abstract
The cardiac work-loop technique closely mimics the intrinsic in vivo movement and characteristics of cardiac muscle function. In this study, six known inotropes were profiled using the work-loop technique to evaluate the potential of this method to predict inotropy. Papillary muscles from male Sprague-Dawley rats were mounted onto an organ bath perfused with Krebs-Henseleit buffer. Following optimisation, work-loop contractions were performed that included an initial stabilisation period followed by vehicle control or drug administration. Six known inotropes were tested: digoxin, dobutamine, isoprenaline, flecainide, verapamil and atenolol. Muscle performance was evaluated by calculating power output during work-loop contraction. Digoxin, dobutamine and isoprenaline caused a significant increase in power output of muscles when compared to vehicle control. Flecainide, verapamil and atenolol significantly reduced power output of muscles. These changes in power output were reflected in alterations in work loop shapes. This is the first study in which changes in work-loop shape detailing for example the activation, shortening or passive re-lengthening have been linked to the mechanism of action of a compound. This study has demonstrated that the work-loop technique can provide an important novel method with which to assess detailed mechanisms of drug-induced effects on cardiac muscle contractility.
Collapse
Affiliation(s)
- Sophie Fletcher
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom.,InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK
| | - Helen Maddock
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom. .,InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK.
| | - Rob S James
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Rob Wallis
- InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK
| | - Mayel Gharanei
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom.,InoCardia Ltd, Technocentre, Puma Way, Coventry, CV1 2TT, UK
| |
Collapse
|
42
|
Early kinetic profiles of troponin I and T measured by high-sensitivity assays in patients with myocardial infarction. Clin Chim Acta 2020; 505:15-25. [PMID: 32061575 DOI: 10.1016/j.cca.2020.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
The early concentration kinetic profiles of cardiac troponin in patients with non-ST-elevated myocardial infarction (NSTEMI) measured by high-sensitivity cardiac troponin I (hs-cTnI) and T (hs-cTnT) assays have not been described. In intermediate-to-high-risk of NSTEMI patients we measured serial cTn concentrations on ED arrival, at 1, 2, 3, 6-12, 24 and 48-hours with hs-cTnI and hs-cTnT assays. Log-normal curves were fitted to concentrations from time from symptom onset, and the time to rule-out decision thresholds estimated (hs-cTnI: 2 ng/L and 5 ng/L; hs-cTnT: 5 ng/L). Among 164 patients there were 58 NSTEMI. The hs-cTnI to hs-cTnT ratio increased linearly over the first 6-12 h following symptom onset. The estimated times from symptom onset to the 2 ng/L and 5 ng/L thresholds for hs-cTnI were 1.8 (0.1-3.1) and 1.9 (1.1-3.5) hours, and to the 5 ng/L threshold for hs-cTnT 1.9 (1.1-3.8) hours. The estimated time to exceed 5 ng/L was ≥3 hours in 32.6% (95%CI: 20.0% to 48.1%) cases for hs-cTnI and 33.3% (19.6% to 50.0%) for hs-cTnT. cTnI concentrations increased at a much more rapid rate than cTnT concentrations in patients with NSTEMI. Concentrations of a high proportion of patients took longer than 3 hours from symptom onset to exceed the 5 ng/L rule-out decision threshold.
Collapse
|
43
|
Cardiac pathology in neuronal ceroid lipofuscinoses (NCL): More than a mere co-morbidity. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165643. [PMID: 31863828 DOI: 10.1016/j.bbadis.2019.165643] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are mostly seen as diseases affecting the central nervous system, but there is accumulating evidence that they have co-morbidities outside the brain. One of these co-morbidities is a decline in cardiac function. This is becoming increasingly recognised in teenagers and adolescents with juvenile CLN3, but it may also occur in individuals with other NCLs. The purpose of this review is to summarise the current knowledge of the structural and functional changes found in the hearts of animal models and people diagnosed with NCL. In addition, we present evidence of structural changes that were observed in a systematic comparison of the cardiomyocytes from CLN3Δex7/8 mice.
Collapse
|
44
|
Pourafkari L, Tajlil A, Nader ND. Biomarkers in diagnosing and treatment of acute heart failure. Biomark Med 2019; 13:1235-1249. [PMID: 31580155 DOI: 10.2217/bmm-2019-0134] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Acute heart failure (AHF) is a complex disorder involving different pathophysiological pathways. In recent years, there is an increased focus on biomarkers that help with diagnosis, risk stratification and disease monitoring of AHF. Finding a reliable set of biomarkers not only improves morbidity and mortality but it can also potentially reveal the new targets of therapy. In this paper, we have reviewed the biomarkers found useful for the diagnosis as well as for risk stratification and prognostication in patients with AHF. We have discussed the established biomarkers for AHF including cardiac troponins and natriuretic peptides and emerging biomarkers including adiponectin, mi-RNA, sST2, Gal-3, MR-proADM, OPG, CT-proAVP and H-FABP for the purposes of making diagnosis, their use as a guide of therapy or for determination of prognosis.
Collapse
Affiliation(s)
- Leili Pourafkari
- Department of Anesthesiology, University at Buffalo, Buffalo, NY 14203, USA
| | - Arezou Tajlil
- Cardiovascular Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
45
|
Sternberg M, Pasini E, Chen-Scarabelli C, Corsetti G, Patel H, Linardi D, Onorati F, Faggian G, Scarabelli T, Saravolatz L. Elevated Cardiac Troponin in Clinical Scenarios Beyond Obstructive Coronary Artery Disease. Med Sci Monit 2019; 25:7115-7125. [PMID: 31542787 PMCID: PMC6774266 DOI: 10.12659/msm.915830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In this systematic review article, we aim to summarize the most up-to-date evidence regarding elevations of cardiac troponin, especially in clinical scenarios other than obstructive coronary artery disease. The accurate interpretation of raised cardiac troponin is challenging because it relies on unconfirmed postulations and dogmatic knowledge (e.g., the exclusive provenience of cardiac troponin from cardiac myocytes), based on which every troponin elevation is assumed to definitely indicate myocardial damage. Indeed, the investigation of the pathophysiologic mechanism leading to the release in the bloodstream of cardiac biomarkers should be the first step of the diagnostic process to fully understand the clinical significance of the elevated serum levels and identify the best management. A prominent effort should be put in place to identify the contribution of potential confounding factors, both cardiac and non-cardiac in etiology, with the ability to affect synthesis and clearance of cardiac biomarkers. Regardless of the underlying cause, it is well established that cardiovascular biomarkers are increasingly useful to further risk stratification and prognosticate patients. Accordingly, we sought to clarify the meaning and impact of elevated cardiac troponin in those frequently encountered real-world scenarios presenting clinicians with a diagnostic dilemma, with the final goal of facilitating the diagnosis and help optimize individually tailored treatment strategies.
Collapse
Affiliation(s)
- Michael Sternberg
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Evasio Pasini
- Scientific Clinical Institutes Maugeri, Cardiac Rehabilitation Lumezzane Institute, Brescia, Italy
| | - Carol Chen-Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| | - Giovannii Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Hemang Patel
- General Medical Education, Department of Internal Medicine, Ascension St. John Hospital, Detroit, MI, USA
| | - Daniele Linardi
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Francesco Onorati
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Giuseppe Faggian
- Division of Cardiovascular Surgery, Verona University Hospital, Verona, Italy
| | - Tiziano Scarabelli
- Center for Heart and Vessel Preclinical Studies, Department of Internal Medicine, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, USA
| | - Louis Saravolatz
- Department of Medicine, Ascension St. John Hospital and Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
46
|
Bowman JD, Lindert S. Computational Studies of Cardiac and Skeletal Troponin. Front Mol Biosci 2019; 6:68. [PMID: 31448287 PMCID: PMC6696891 DOI: 10.3389/fmolb.2019.00068] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022] Open
Abstract
Troponin is a key regulatory protein in muscle contraction, consisting of three subunits troponin C (TnC), troponin I (TnI), and troponin T (TnT). Calcium association to TnC initiates contraction by causing a series of dynamic and conformational changes that allow the switch peptide of TnI to bind and subsequently cross bridges to form between the thin and thick filament of the sarcomere. Owing to its pivotal role in contraction regulation, troponin has been the focus of numerous computational studies over the last decade. These studies elegantly supplemented a large volume of experimental work and focused on the structure, dynamics and function of the whole troponin complex, individual subunits, and even on segments of the thin filament. Molecular dynamics, Brownian dynamics, and free energy simulations have been used to elucidate the conformational dynamics and underlying free energy landscape of troponin, calcium, and switch peptide binding, as well as the effect of disease mutations, small molecules and post-translational modifications such as phosphorylation. Frequently, simulations have been used to confirm or explain experimental observations. Computer-aided drug discovery tools have been employed to identify novel potential calcium sensitizing agents binding to the TnC-TnI interface. Finally, Markov modeling has contributed to simulating contraction within the sarcomere on the mesoscale. Here we are reviewing and classifying the existing computational work on troponin and its subunits, outline current gaps in simulations elucidating troponin's role in contraction and suggest potential future developments in the field.
Collapse
Affiliation(s)
- Jacob D Bowman
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| |
Collapse
|
47
|
Lieben Louis X, Raj P, Meikle Z, Yu L, Susser SE, MacInnis S, Duhamel TA, Wigle JT, Netticadan T. Resveratrol prevents palmitic-acid-induced cardiomyocyte contractile impairment. Can J Physiol Pharmacol 2019; 97:1132-1140. [PMID: 31374178 DOI: 10.1139/cjpp-2019-0051] [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: 12/16/2022]
Abstract
Long-chain saturated fatty acids, especially palmitic acid (PA), contribute to cardiomyocyte lipotoxicity. This study tests the effects of PA on adult rat cardiomyocyte contractile function and proteins associated with calcium regulating cardiomyocyte contraction and relaxation. Adult rat cardiomyocytes were pretreated with resveratrol (Resv) and then treated with PA. For the reversal study, cardiomyocytes were incubated with PA prior to treatment with Resv. Cardiomyocyte contractility, ratio of rod- to round-shaped cardiomyocytes, and Hoechst staining were used to measure functional and morphological changes in cardiomyocytes. Protein expression of sarco-endoplasmic reticulum ATPase 2a (SERCA2a), native phospholamban (PLB) and phosphorylated PLB (pPLB ser16 and pPLB thr17), and troponin I (TnI) and phosphorylated TnI (pTnI) were measured. SERCA2a activity was also measured. Our results show that PA (200 μM) decreased the rate of cardiomyocyte relaxation, reduced the number of rod-shaped cardiomyocytes, and increased the number of cells with condensed nuclei; pre-treating cardiomyocytes with Resv significantly prevented these changes. Post-treatment with Resv did not reverse morphological changes induced by PA. Protein expression levels of SERCA2a, PLB, pPLBs, TnI, and pTnI were unchanged by PA or Resv. SERCA2a activity assay showed that Vmax and Iono ratio were increased with PA and pre-treatment with Resv prevented this increase. In conclusion, our results show that Resv protect cardiomyocytes from contractile dysfunction induced by PA.
Collapse
Affiliation(s)
- Xavier Lieben Louis
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen, Research Centre, Winnipeg, MB R2H 2A6, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Pema Raj
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen, Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Zach Meikle
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Liping Yu
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen, Research Centre, Winnipeg, MB R2H 2A6, Canada.,Agriculture and Agri-Food Canada, Winnipeg, MB R2H 2A6, Canada
| | - Shannel E Susser
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Shayla MacInnis
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Todd A Duhamel
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,Health, Leisure & Human Performance Research Institute, Faculty of Kinesiology & Recreation Management, University of Manitoba, MB R3E 0J9, Canada
| | - Jeffrey T Wigle
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Thomas Netticadan
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen, Research Centre, Winnipeg, MB R2H 2A6, Canada.,Agriculture and Agri-Food Canada, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
48
|
Adler C, Baldus S. [Troponin elevation-does every patient require coronary angiography?]. Med Klin Intensivmed Notfmed 2019; 115:690-698. [PMID: 31218391 DOI: 10.1007/s00063-019-0593-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 04/29/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023]
Abstract
The determination of high-sensitivity troponin remains the gold standard in patients with acute chest pain and allows the early diagnosis and therapy of patients with acute myocardial infarction (AMI). Increased cardiac troponin levels are highly specific for myocardial damage. However, elevated troponin levels are not equivalent to AMI and therefore the clinical presentation and the electrocardiogram of each patient must be taken into account. Elevated troponin levels are found in numerous diseases but not every patient needs invasive coronary angiography.
Collapse
Affiliation(s)
- Christoph Adler
- Klinik für Kardiologie, Angiologie, Pneumologie und internistische Intensivmedizin, Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - Stephan Baldus
- Klinik für Kardiologie, Angiologie, Pneumologie und internistische Intensivmedizin, Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| |
Collapse
|
49
|
Viola HM, Hool LC. Impaired calcium handling and mitochondrial metabolic dysfunction as early markers of hypertrophic cardiomyopathy. Arch Biochem Biophys 2019; 665:166-174. [PMID: 30885674 DOI: 10.1016/j.abb.2019.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder, characterised by myocyte remodeling, disorganisation of sarcomeric proteins, impaired energy metabolism and altered cardiac contractility. Gene mutations encoding cardiac contractile proteins account for 60% of HCM aetiology. Current drug therapy including L-type calcium channel antagonists, are used to manage symptoms in patients with overt HCM, but no treatment exists that can reverse or prevent the cardiomyopathy. Design of effective drug therapy will require a clear understanding of the early pathophysiological mechanisms of the disease. Numerous studies have investigated specific aspects of HCM pathophysiology. This review brings these findings together, in order to develop a holistic understanding of the early pathophysiological mechanisms of the disease. We focus on gene mutations in cardiac myosin binding protein-C, β-cardiac myosin heavy chain, cardiac troponin I, and cardiac troponin T, that comprise the majority of all HCM sarcomeric gene mutations. We find that although some similarities exist, each mutation leads to mutation-specific alterations in calcium handling, myofilament calcium sensitivity and mitochondrial metabolic function. This may contribute to the observed clinical phenotypic variability in sarcomeric-related HCM. An understanding of early mutation-specific mechanisms of the disease may provide useful markers of disease progression, and inform therapeutic design.
Collapse
Affiliation(s)
- Helena M Viola
- School of Human Sciences (Physiology), The University of Western Australia, Crawley, WA, Australia
| | - Livia C Hool
- School of Human Sciences (Physiology), The University of Western Australia, Crawley, WA, Australia; Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
50
|
Romiti GF, Cangemi R, Toriello F, Ruscio E, Sciomer S, Moscucci F, Vincenti M, Crescioli C, Proietti M, Basili S, Raparelli V. Sex-Specific Cut-Offs for High-Sensitivity Cardiac Troponin: Is Less More? Cardiovasc Ther 2019; 2019:9546931. [PMID: 31772621 PMCID: PMC6739766 DOI: 10.1155/2019/9546931] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
Management of patients presenting to the Emergency Department with chest pain is continuously evolving. In the setting of acute coronary syndrome, the availability of high-sensitivity cardiac troponin assays (hs-cTn) has allowed for the development of algorithms aimed at rapidly assessing the risk of an ongoing myocardial infarction. However, concerns were raised about the massive application of such a simplified approach to heterogeneous real-world populations. As a result, there is a potential risk of underdiagnosis in several clusters of patients, including women, for whom a lower threshold for hs-cTn was suggested to be more appropriate. Implementation in clinical practice of sex-tailored cut-off values for hs-cTn represents a hot topic due to the need to reduce inequality and improve diagnostic performance in females. The aim of this review is to summarize current evidence on sex-specific cut-off values of hs-cTn and their application and usefulness in clinical practice. We also offer an extensive overview of thresholds reported in literature and of the mechanisms underlying such differences among sexes, suggesting possible explanations about debated issues.
Collapse
Affiliation(s)
- Giulio Francesco Romiti
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Roberto Cangemi
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Filippo Toriello
- Division of Cardiology, San Paolo Hospital, Department of Health Sciences, University of Milan, Milan, Italy
| | - Eleonora Ruscio
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Susanna Sciomer
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza–University of Rome, Rome, Italy
| | - Federica Moscucci
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza–University of Rome, Rome, Italy
| | - Marianna Vincenti
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Marco Proietti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Stefania Basili
- Department of Internal Medicine and Medical Specialties, Sapienza–University of Rome, Rome, Italy
| | - Valeria Raparelli
- Department of Experimental Medicine, Sapienza–University of Rome, Rome, Italy
- Center for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|