51
|
Deep Learning Analyses to Delineate the Molecular Remodeling Process after Myocardial Infarction. Cells 2021; 10:cells10123268. [PMID: 34943776 PMCID: PMC8699769 DOI: 10.3390/cells10123268] [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: 10/13/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 02/01/2023] Open
Abstract
Specific proteins and processes have been identified in post-myocardial infarction (MI) pathological remodeling, but a comprehensive understanding of the complete molecular evolution is lacking. We generated microarray data from swine heart biopsies at baseline and 6, 30, and 45 days after infarction to feed machine-learning algorithms. We cross-validated the results using available clinical and experimental information. MI progression was accompanied by the regulation of adipogenesis, fatty acid metabolism, and epithelial-mesenchymal transition. The infarct core region was enriched in processes related to muscle contraction and membrane depolarization. Angiogenesis was among the first morphogenic responses detected as being sustained over time, but other processes suggesting post-ischemic recapitulation of embryogenic processes were also observed. Finally, protein-triggering analysis established the key genes mediating each process at each time point, as well as the complete adverse remodeling response. We modeled the behaviors of these genes, generating a description of the integrative mechanism of action for MI progression. This mechanistic analysis overlapped at different time points; the common pathways between the source proteins and cardiac remodeling involved IGF1R, RAF1, KPCA, JUN, and PTN11 as modulators. Thus, our data delineate a structured and comprehensive picture of the molecular remodeling process, identify new potential biomarkers or therapeutic targets, and establish therapeutic windows during disease progression.
Collapse
|
52
|
Aldosterone Blockade in Acute Myocardial Infarction: A Systematic Review and Meta-Analysis. Cardiovasc Ther 2021; 2021:1710731. [PMID: 34786023 PMCID: PMC8560277 DOI: 10.1155/2021/1710731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/02/2021] [Accepted: 09/18/2021] [Indexed: 12/12/2022] Open
Abstract
Background A comprehensive evaluation of the benefits of mineralocorticoid receptor antagonists (MRA) in acute myocardial infarction (AMI) patients is lacking. Objective To summarize the evidence on the efficacy and safety of MRA in patients admitted for AMI. Methods Articles were identified through PubMed, Embase, Cochrane Library, Ovid (Medline1946-2021), and ClinicalTrials.gov databases from their inception to December 31, 2020. Results 15 articles with a total of 11,861 patients were included. MRA reduced the risk of all-cause mortality by 16% (relative ratio (RR): 0.84; 95% confidence interval (CI) (0.76, 0.94); P = 0.002) and the incidence of cardiovascular adverse events by 12% (RR: 0.88, 95% CI (0.83, 0.93), P < 0.00001) in post-AMI patients, and further analysis demonstrated that early administration of MRA within 7 days after AMI resulted in a greater reduction in all-cause mortality (RR: 0.72, 95% CI (0.61, 0.85), P < 0.0001). Subgroup analyses showed that post-STEMI patients without left ventricular systolic dysfunction (LVSD) treated with MRA had a 36% reduction in all-cause mortality (RR: 0.64, 95% CI (0.46, 0.89), P = 0.007) and a 22% reduction in cardiovascular adverse events (RR: 0.78, 95% CI (0.67, 0.91), P = 0.002). Meanwhile, post-STEMI patients without LVSD treated with MRA get significant improvements in left ventricular ejection fraction (mean difference (MD): 2.69, 95% CI (2.44, 2.93), P < 0.00001), left ventricular end-systolic index (MD: -4.52 ml/m2, 95% CI (-8.21, -0.83), P = 0.02), and left ventricular end-diastolic diameter (MD: -0.11 cm, 95% CI (-0.22, 0.00), P = 0.05). The corresponding RR were 1.72 (95% CI (1.43, 2.07), P < 0.00001) for considered common adverse events (hyperkalemia, gynecomastia, and renal dysfunction). Conclusions Our findings suggest that MRA treatment reduces all-cause mortality and cardiovascular adverse events in post-AMI patients, which is more significant in patients after STEMI without LVSD. In addition, MRA treatment may exert beneficial effects on the reversal of cardiac remodeling in patients after STEMI without LVSD.
Collapse
|
53
|
Chen XY, Yang QM, Fang JX, Guo HF. The Value of Three-Dimensional Speckle-Tracking Echocardiography in the Prediction of Cardiovascular Events in Patients with Hypertension Complicated by Acute Myocardial Infarction: A Long-Term Follow-Up Study. Int J Gen Med 2021; 14:6703-6712. [PMID: 34675631 PMCID: PMC8520435 DOI: 10.2147/ijgm.s330789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
Background Patients with hypertension complicated by acute myocardial infarction (AMI) have a generally poor prognosis. The identification of powerful predictors for recurring cardiovascular events (RCEs) is vital. This study seeks to evaluate the predictive value of three-dimensional (3D) strain parameters for RCEs in patients with hypertension complicated by AMI. Methods We successfully followed up patients with hypertension and AMI from April 2015 to December 2015 in this retrospective study. Participants previously underwent 3D echocardiography, one week, and one month after percutaneous coronary intervention (PCI). The left ventricular structural function parameters, as well as 3D strain parameters including 3D global longitudinal strain (3D-GLS), 3D global circumferential strain, 3D global radial strain, and 3D global area strain (3D-GAS), were acquired. A Cox model was used to determine the relationships between these parameters and RCEs. Results In total, 62 patients were enrolled in the analysis. During follow-up (41.27 ± 20.45 months), 20 patients (32.3%) had RCEs that were independently predicted one month after PCI by 3D-GLS (HR: 1.481, 95% CI: 1.202–1.824, P < 0.001) and 3D-GAS (HR: 1.254, 95% CI: 1.093–1.440, P = 0.001). The optimal cutoffs for 3D-GLS and 3D-GAS in predicting cardiac events were ≥12.5% (area under the receiver operating characteristic curve [AUC]: 0.736, 95% CI: 0.611–0.862, P = 0.003), and >20.5% (AUC: 0.685, 95% CI: 0.551–0.818, P = 0.020), respectively. Conclusion The assessed values of 3D-GLS and 3D-GAS one month after PCI can predict RCEs in patients with hypertension complicated by AMI.
Collapse
Affiliation(s)
- Xiao-Yan Chen
- Department of Ultrasound, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.,Department of Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Qing-Mei Yang
- Department of Ultrasound, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.,Department of Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jian-Xiu Fang
- Department of Ultrasound, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.,Department of Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Hai-Feng Guo
- Department of Ultrasound, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.,Department of Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| |
Collapse
|
54
|
Xiong W, Feng S, Wang H, Qing S, Yang Y, Zhao Y, Zeng Z, Gong J. Identification of candidate genes and pathways in limonin-mediated cardiac repair after myocardial infarction. Biomed Pharmacother 2021; 142:112088. [PMID: 34470729 DOI: 10.1016/j.biopha.2021.112088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/24/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Myocardial infarction (MI) resulting from acute coronary ischemia may cause significant morbidity and mortality, and microRNAs play a vital role in this pathophysiology. Limonin (LIM) is a natural medicine from citrus fruit that protects organs against ischemic diseases, but the candidate genes and pathways associated with cardioprotection are unknown. METHODS MI was induced by ligating the left anterior descending coronary in male Sprague-Dawley rats. LIM was orally administered for 7 days after the induction of MI. Subsequently, the hearts were collected to examine significant changes in microRNAs and mRNAs among the control (CON), MI, and LIM + MI groups. Gene Ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and protein-protein interaction (PPI) networks were used to identify the biological functions and signaling pathways of differentially expressed mRNAs. Candidate genes were validated by RT-qPCR. RESULTS Compared to the CON group, MI caused significant changes in the expression of 26 microRNAs and 1979 mRNAs. The bioinformatics analysis showed that inflammation, apoptosis, and oxidation were enriched in GO terms, while RAP1, PI3K/AKT, RAS, and cGMP-PKG were enriched in KEGG pathways. In addition, compared to the MI group, LIM induced significant changes in the expression of 4 microRNAs and 173 mRNAs. The differentially expressed mRNAs were related to collagen biosynthesis, the immune response, extrinsic apoptosis, and tight junctions. One microRNA (rno-miR-10a-5p) and 2 mRNAs (IGLON5 and LMX1A) were differentially expressed among the CON, MI, and LIM + MI groups. CONCLUSIONS Our results suggest that the rno-miR-10a-5p-IGLON5/LMX1A axis may be a candidate pathway and promising target through which LIM alleviates MI-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China
| | - Shiyan Feng
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China; Emergency Medical Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, Sichuan, China
| | - Hong Wang
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China; Department of Emergency Intensive Care Unit, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China
| | - Song Qing
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China
| | - Yong Yang
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China; Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, Sichuan, China
| | - Yanhua Zhao
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China
| | - Zhongbo Zeng
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China
| | - Jian Gong
- Department of Clinical Research, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China; Department of Emergency Intensive Care Unit, Ziyang People's Hospital, Ziyang Hospital of Sichuan Provincial People's Hospital, Ziyang, Sichuan, China.
| |
Collapse
|
55
|
Flores-Vergara R, Olmedo I, Aránguiz P, Riquelme JA, Vivar R, Pedrozo Z. Communication Between Cardiomyocytes and Fibroblasts During Cardiac Ischemia/Reperfusion and Remodeling: Roles of TGF-β, CTGF, the Renin Angiotensin Axis, and Non-coding RNA Molecules. Front Physiol 2021; 12:716721. [PMID: 34539441 PMCID: PMC8446518 DOI: 10.3389/fphys.2021.716721] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022] Open
Abstract
Communication between cells is a foundational concept for understanding the physiology and pathology of biological systems. Paracrine/autocrine signaling, direct cell-to-cell interplay, and extracellular matrix interactions are three types of cell communication that regulate responses to different stimuli. In the heart, cardiomyocytes, fibroblasts, and endothelial cells interact to form the cardiac tissue. Under pathological conditions, such as myocardial infarction, humoral factors released by these cells may induce tissue damage or protection, depending on the type and concentration of molecules secreted. Cardiac remodeling is also mediated by the factors secreted by cardiomyocytes and fibroblasts that are involved in the extensive reciprocal interactions between these cells. Identifying the molecules and cellular signal pathways implicated in these processes will be crucial for creating effective tissue-preserving treatments during or after reperfusion. Numerous therapies to protect cardiac tissue from reperfusion-induced injury have been explored, and ample pre-clinical research has attempted to identify drugs or techniques to mitigate cardiac damage. However, despite great success in animal models, it has not been possible to completely translate these cardioprotective effects to human applications. This review provides a current summary of the principal molecules, pathways, and mechanisms underlying cardiomyocyte and cardiac fibroblast crosstalk during ischemia/reperfusion injury. We also discuss pre-clinical molecules proposed as treatments for myocardial infarction and provide a clinical perspective on these potential therapeutic agents.
Collapse
Affiliation(s)
- Raúl Flores-Vergara
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Ivonne Olmedo
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago de Chile, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, Viña del Mar, Chile
| | - Jaime Andrés Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago de Chile, Chile
| | - Raúl Vivar
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Zully Pedrozo
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago de Chile, Chile
| |
Collapse
|
56
|
Ferlazzo N, Currò M, Isola G, Maggio S, Bertuccio MP, Trovato-Salinaro A, Matarese G, Alibrandi A, Caccamo D, Ientile R. Changes in the Biomarkers of Oxidative/Nitrosative Stress and Endothelial Dysfunction Are Associated with Cardiovascular Risk in Periodontitis Patients. Curr Issues Mol Biol 2021; 43:704-715. [PMID: 34287264 PMCID: PMC8929118 DOI: 10.3390/cimb43020051] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with cardiovascular disease (CVD) and periodontitis (PT) show shared risk factors as result of the altered molecular mechanisms associated with pathological conditions. The aim of our study was to evaluate if the plasma biomarkers associated with endothelial dysfunction may also be related to alterations in the inflammatory status in peripheral blood mononuclear cells (PBMC). Patients with PT, coronary heart disease (CHD), or both diseases as well as controls were enrolled. Plasma levels of coenzyme Q10 (CoQ10), 3-nitrotyrosine (NT), and asymmetric dimethylarginine (ADMA) were assessed using HPLC. mRNA levels of caspase-1 (CASP1), NLR family pyrin domain containing 3 (NLRP3), and tumor necrosis factor-α (TNF-α) in PBMC from the recruited subjects were quantified using real-time PCR. Patients with PT + CHD showed lower CoQ10 plasma levels and increased concentrations of NT in comparison to healthy subjects. ADMA levels were higher in CHD and PT + CHD patients compared to controls. Transcript levels of CASP1, NLRP3, and TNF-α were up-regulated in PBMC from all patient groups when compared to healthy subjects. Our results suggest a possible causal link between oxidative stress, high levels of NT and ADMA, and inflammasome activation, which may be involved in the endothelial inflammatory dysfunction leading to the pathogenesis and progression of CHD in PT patients.
Collapse
Affiliation(s)
- Nadia Ferlazzo
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
- Correspondence: (N.F.); (M.C.); Tel.: +39-0902213389 (M.C.)
| | - Monica Currò
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
- Correspondence: (N.F.); (M.C.); Tel.: +39-0902213389 (M.C.)
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via S. Sofia 78, 95123 Catania, Italy;
| | - Silvia Maggio
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | - Maria Paola Bertuccio
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | | | - Giovanni Matarese
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | - Angela Alibrandi
- Unit of Statistical and Mathematical Sciences, Department of Economics, University of Messina, 98123 Messina, Italy;
| | - Daniela Caccamo
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | - Riccardo Ientile
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| |
Collapse
|
57
|
Tilea I, Varga A, Serban RC. Past, Present, and Future of Blood Biomarkers for the Diagnosis of Acute Myocardial Infarction-Promises and Challenges. Diagnostics (Basel) 2021; 11:diagnostics11050881. [PMID: 34063483 PMCID: PMC8156776 DOI: 10.3390/diagnostics11050881] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/28/2022] Open
Abstract
Despite important advancements in acute myocardial infarction (AMI) management, it continues to represent a leading cause of mortality worldwide. Fast and reliable AMI diagnosis can significantly reduce mortality in this high-risk population. Diagnosis of AMI has relied on biomarker evaluation for more than 50 years. The upturn of high-sensitivity cardiac troponin testing provided extremely sensitive means to detect cardiac myocyte necrosis, but this increased sensitivity came at the cost of a decrease in diagnostic specificity. In addition, although cardiac troponins increase relatively early after the onset of AMI, they still leave a time gap between the onset of myocardial ischemia and our ability to detect it, thus precluding very early management of AMI. Newer biomarkers detected in processes such as inflammation, neurohormonal activation, or myocardial stress occur much earlier than myocyte necrosis and the diagnostic rise of cardiac troponins, allowing us to expand biomarker research in these areas. Increased understanding of the complex AMI pathophysiology has spurred the search of new biomarkers that could overcome these shortcomings, whereas multi-omic and multi-biomarker approaches promise to be game changers in AMI biomarker assessment. In this review, we discuss the evolution, current application, and emerging blood biomarkers for the diagnosis of AMI; we address their advantages and promises to improve patient care, as well as their challenges, limitations, and technical and diagnostic pitfalls. Questions that remain to be answered and hotspots for future research are also emphasized.
Collapse
Affiliation(s)
- Ioan Tilea
- Department M4, Clinical Sciences, Faculty of Medicine, “G. E. Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Department of Cardiology II, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
| | - Andreea Varga
- Department of Cardiology II, Emergency Clinical County Hospital, 540042 Targu Mures, Romania
- Department ME2, Faculty of Medicine in English, “G. E. Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- Correspondence: ; Tel.: +40-730808111
| | - Razvan Constantin Serban
- Cardiac Catheterization Laboratory, The Emergency Institute for Cardiovascular Diseases and Transplantation, 540136 Targu Mures, Romania;
| |
Collapse
|
58
|
Testa A, Frati G, Versaci F, Biondi-Zoccai G. Ischemia-Reperfusion Injury: Can We Stop the Curing-Hurting Paradox? J Cardiovasc Pharmacol 2021; 77:427-429. [PMID: 33818549 DOI: 10.1097/fjc.0000000000000992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Alberto Testa
- Sapienza School for Advanced Studies, Sapienza University of Rome, Rome, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS NEUROMED, Pozzilli, Italy
| | - Francesco Versaci
- Unità Operativa Complessa di Cardiologia, Ospedale Santa Maria Goretti, Latina, Italy; and
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| |
Collapse
|
59
|
Węgiel M, Rakowski T. Circulating biomarkers as predictors of left ventricular remodeling after myocardial infarction. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2021; 17:21-32. [PMID: 33868414 PMCID: PMC8039920 DOI: 10.5114/aic.2021.104764] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/19/2020] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The main impact of myocardial infarction is shifting from acute mortality to adverse remodeling and chronic left ventricle dysfunction. Several circulating biomarkers are explored for better risk stratification of these patients. Biomarker testing is a very attractive idea, since it is non-invasive, not operator-dependent and widely available. AIM In the present paper we analyze data from the years 2005-2020 about circulating biomarkers of remodeling after myocardial infarction. MATERIAL AND METHODS We assessed 53 articles, which examined 160 relations between biomarkers and remodeling. We analyze inclusion criteria for individual studies, time points of serum collection and remodeling assessment as well as imaging methods. RESULTS The main groups of assessed biomarkers included B-type natriuretic peptides, markers of cardiomyocyte injury and necrosis, markers of inflammatory response, markers of extracellular matrix turnover, microRNAs and hormones. The most common method of remodeling assessment was echocardiography and the most frequent time point for remodeling evaluation was 6 months. CONCLUSIONS The present analysis shows that although a relatively large number biomarkers were tested, selecting one ideal marker is still a challenge. A combination of biomarkers from different groups might be appropriate for predicting remodeling. Data presented in this analysis might be helpful for designing future studies, evaluating clinical use of an individual biomarker or a combination of different biomarkers.
Collapse
Affiliation(s)
- Michał Węgiel
- 2 Department of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz Rakowski
- 2 Department of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
60
|
Chen Q, Yin Q, Song J, Liu C, Chen H, Li S. Identification of monocyte-associated genes as predictive biomarkers of heart failure after acute myocardial infarction. BMC Med Genomics 2021; 14:44. [PMID: 33563285 PMCID: PMC7871627 DOI: 10.1186/s12920-021-00890-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/31/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a major contributor of heart failure (HF). Peripheral blood mononuclear cells (PBMCs), mainly monocytes, are the essential initiators of AMI-induced HF. The powerful biomarkers for early identification of AMI patients at risk of HF remain elusive. We aimed to identify monocyte-related critical genes as predictive biomarkers for post-AMI HF. METHODS We performed weighted gene co-expression network analysis (WGCNA) on transcriptomics of PBMCs from AMI patients who developed HF or did not. Functional enrichment analysis of genes in significant modules was performed via Metascape. Then we obtained the single-cell RNA-sequencing data of recruited monocytes/macrophages from AMI and control mice using the Scanpy and screened 381 differentially expressed genes (DEGs) between the two groups. We validated the expression changes of the 25 genes in cardiac macrophages from AMI mice based on bulk RNA-sequencing data and PBMCs data mentioned above. RESULTS In our study, the results of WGCNA showed that two modules containing 827 hub genes were most significantly associated with post-AMI HF, which mainly participated in cell migration, inflammation, immunity, and apoptosis. There were 25 common genes between DEGs and hub genes, showing close relationship with inflammation and collagen metabolism. CUX1, CTSD and ADD3 exhibited consistent changes in three independent studies. Receiver operating characteristic curve analysis showed that each of the three genes had excellent performance in recognizing post-AMI HF patients. CONCLUSION Our findings provided a set of three monocyte-related biomarkers for the early prediction of HF development after AMI as well as potential therapeutic targets of post-AMI HF.
Collapse
Affiliation(s)
- Qixin Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Qijin Yin
- Ministry of Education Key Laboratory of Bioinformatics, Research Department of Bioinformatics at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Junxian Song
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Chuanfen Liu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Hong Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| | - Sufang Li
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, No 11. Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
61
|
Cheong CS, Aung AT, Wong RC, Yeo TJ, Chan SP, Lee CH. Obstructive sleep apnea during acute coronary syndrome is related to myocardial necrosis and wall stress. Sleep Med 2021; 79:79-82. [PMID: 33482456 DOI: 10.1016/j.sleep.2021.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/26/2020] [Accepted: 01/03/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The relative contribution of pathophysiological mechanisms in acute coronary syndrome (ACS) towards obstructive sleep apnea (OSA) is not well-studied. We examined the correlation between severity of OSA and inflammation, myocardial necrosis, wall stress, and fibrosis. METHODS A total of 89 patients admitted with ACS underwent a sleep study during index admission. Plasma levels of high-sensitivity C-reactive protein (hs-CRP), troponin I, N-terminal pro-brain natriuretic peptide (NT-proBNP), and suppression of tumorigenicity 2 (ST2) were prospectively analyzed. Two patients diagnosed with central sleep apnea were excluded. RESULTS The recruited patients were divided into no (AHI <5 events/hour, 9.2%), mild (5-<15, 27.6%), moderate (15-<30, 21.8%), and severe (≥30, 41.4%) OSA. Compared to the no, mild and moderate OSA groups, the severe OSA group had a higher body mass index (p = 0.005). They were also more likely to present with ST-segment elevation ACS (versus non-ST-segment elevation ACS) (p = 0.041), have undergone previous coronary artery bypass grafting (p = 0.013), demonstrate complete coronary occlusion during baseline coronary angiography (p = 0.049), and have a larger left atrial diameter measured on echocardiography (p = 0.029). Likewise, the severe OSA group had higher plasma levels of hs-CRP (p = 0.004), troponin I (p = 0.017), and NT-proBNP (p = 0.004), but not ST2 (p = 0.10). After adjustment for the effects of confounding variables, OSA was independently associated with troponin I (ie, myocardial necrosis; p = 0.001) and NT-proBNP (ie, myocardial wall stress; p = 0.008). CONCLUSION Severe OSA during the acute phase of ACS was associated with extensive myocardial necrosis and high myocardial wall stress, but not with inflammation and myocardial fibrosis.
Collapse
Affiliation(s)
- Crystal Sj Cheong
- Department of Otolaryngology - Head & Neck Surgery, National University Hospital, Singapore
| | - Aye-Thandar Aung
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Raymond Cc Wong
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Tee Joo Yeo
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Siew-Pang Chan
- Department of Cardiology, National University Heart Centre, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Research Institute, National University of Singapore, Singapore
| | - Chi-Hang Lee
- Department of Cardiology, National University Heart Centre, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Research Institute, National University of Singapore, Singapore.
| |
Collapse
|
62
|
Bostan MM, Stătescu C, Anghel L, Șerban IL, Cojocaru E, Sascău R. Post-Myocardial Infarction Ventricular Remodeling Biomarkers-The Key Link between Pathophysiology and Clinic. Biomolecules 2020; 10:E1587. [PMID: 33238444 PMCID: PMC7700609 DOI: 10.3390/biom10111587] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Studies in recent years have shown increased interest in developing new methods of evaluation, but also in limiting post infarction ventricular remodeling, hoping to improve ventricular function and the further evolution of the patient. This is the point where biomarkers have proven effective in early detection of remodeling phenomena. There are six main processes that promote the remodeling and each of them has specific biomarkers that can be used in predicting the evolution (myocardial necrosis, neurohormonal activation, inflammatory reaction, hypertrophy and fibrosis, apoptosis, mixed processes). Some of the biomarkers such as creatine kinase-myocardial band (CK-MB), troponin, and N-terminal-pro type B natriuretic peptide (NT-proBNP) were so convincing that they immediately found their place in the post infarction patient evaluation protocol. Others that are related to more complex processes such as inflammatory biomarkers, atheroma plaque destabilization biomarkers, and microRNA are still being studied, but the results so far are promising. This article aims to review the markers used so far, but also the existing data on new markers that could be considered, taking into consideration the most important studies that have been conducted so far.
Collapse
Affiliation(s)
- Maria-Madălina Bostan
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (M.-M.B.); (R.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M.Georgescu”, 700503 Iasi, Romania
| | - Cristian Stătescu
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (M.-M.B.); (R.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M.Georgescu”, 700503 Iasi, Romania
| | - Larisa Anghel
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (M.-M.B.); (R.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M.Georgescu”, 700503 Iasi, Romania
| | | | - Elena Cojocaru
- Department of Morphofunctional Sciences I—Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania;
| | - Radu Sascău
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (M.-M.B.); (R.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M.Georgescu”, 700503 Iasi, Romania
| |
Collapse
|