1
|
He J, Kong L, An D, Chen B, Zhao C, Li Z, Yang F, Dong J, Wei L, Shan P, Chen Y, Wu L, Xu J, Ge H, Pu J. Prognostic Value of Segmental Strain After ST-Elevation Myocardial Infarction: Insights From the EARLY Assessment of MYOcardial Tissue Characteristics by Cardiac Magnetic Resonance (EARLY-MYO-CMR) Study. J Magn Reson Imaging 2024; 60:2002-2017. [PMID: 38363170 DOI: 10.1002/jmri.29274] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND The prognostic value of left ventricular segmental strain (SS) in ST-elevation myocardial infarction (STEMI) remains unclear. HYPOTHESIS To assess the prognostic value and application of SS. STUDY TYPE Retrospective analysis of a prospective registry. POPULATION Five hundred and forty-four patients after STEMI (500 in Cohort 1, 44 in Cohort 2). FIELD STRENGTH/SEQUENCE 3 T, balanced steady-state free precession, gradient echo, and gradient echo contrast-enhanced images. ASSESSMENT Participants underwent cardiac MR during the acute phase after STEMI. Infarct-related artery (IRA) strain was determined based on SS obtained from cine images. The primary endpoint was the composite of major adverse cardiovascular events (MACEs) after 8 years of follow-up. In Cohort 2, SS stability was assessed by MR twice within 8 days. Contrast and non-contrast risk models based on SS were established, leading to the development of an algorithm. STATISTICAL TEST Student's t-test, Mann-Whitney U-test, Cox and logistic regression, Kaplan-Meier analysis, net reclassification index (NRI). P < 0.05 was considered significant. RESULTS During a median follow-up of 5.2 years, 83 patients from Cohort 1 experienced a MACE. Among SS, IRA peak circumferential strain (IRA-CS) was an independent factor for MACEs (adjusted hazard ratio 1.099), providing incremental prognostic value (NRI 0.180, P = 0.10). Patients with worse IRA-CS (>-8.64%) demonstrated a heightened susceptibility to MACE. Additionally, IRA-CS was significantly associated with microvascular obstruction (MVO) (adjusted odds ratio 1.084) and infarct size (r = 0.395). IRA-CS showed comparable prognostic effectiveness to global peak circumferential strain (NRI 0.100, P = 0.39), also counterbalancing contrast and non-contrast risk models (NRI 0.205, P = 0.05). In Cohort 2, IRA-CS demonstrated stability between two time points (P = 0.10). Based on risk models incorporating IRA-CS, algorithm "HJKL" was preliminarily proposed for stratification. DATA CONCLUSIONS IRA-CS is an important prognostic factor, and an algorithm based on it is proposed for stratification. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jie He
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingcong Kong
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dongaolei An
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Binghua Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chengxu Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Li
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Yang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianxun Dong
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lai Wei
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peiren Shan
- Department of Cardiology, Wenzhou Medical University Affiliated NO. 1 Hospital, Wenzhou, China
| | - Yingmin Chen
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lianming Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianrong Xu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Heng Ge
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
2
|
Pei Z, Qiu J, Zhao Y, Song S, Wang R, Luo W, Cai X, Liu B, Chen H, Yin J, Weng X, Wu Y, Li C, Shen L, Ge J. A novel intracoronary hypothermia device reduces myocardial reperfusion injury in pigs. Chin Med J (Engl) 2024; 137:2461-2472. [PMID: 38445387 PMCID: PMC11479452 DOI: 10.1097/cm9.0000000000003033] [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: 09/19/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Hypothermia therapy has been suggested to attenuate myocardial necrosis; however, the clinical implementation as a valid therapeutic strategy has failed, and new approaches are needed to translate into clinical applications. This study aimed to assess the feasibility, safety, and efficacy of a novel selective intracoronary hypothermia (SICH) device in mitigating myocardial reperfusion injury. METHODS This study comprised two phases. The first phase of the SICH was performed in a normal porcine model for 30 minutes ( n = 5) to evaluate its feasibility. The second phase was conducted in a porcine myocardial infarction (MI) model of myocardial ischemia/reperfusion which was performed by balloon occlusion of the left anterior descending coronary artery for 60 minutes and maintained for 42 days. Pigs in the hypothermia group ( n = 8) received hypothermia intervention onset reperfusion for 30 minutes and controls ( n = 8) received no intervention. All animals were followed for 42 days. Cardiac magnetic resonance analysis (five and 42 days post-MI) and a series of biomarkers/histological studies were performed. RESULTS The average time to lower temperatures to a steady state was 4.8 ± 0.8 s. SICH had no impact on blood pressure or heart rate and was safely performed without complications by using a 3.9 F catheter. Interleukin-6 (IL-6), tumor necrosis factor-α, C-reactive protein (CRP), and brain natriuretic peptide (BNP) were lower at 60 min post perfusion in pigs that underwent SICH as compared with the control group. On day 5 post MI/R, edema, intramyocardial hemorrhage, and microvascular obstruction were reduced in the hypothermia group. On day 42 post MI/R, the infarct size, IL-6, CRP, BNP, and matrix metalloproteinase-9 were reduced, and the ejection fraction was improved in pigs that underwent SICH. CONCLUSIONS The SICH device safely and effectively reduced the infarct size and improved heart function in a pig model of MI/R. These beneficial effects indicate the clinical potential of SICH for treatment of myocardial reperfusion injury.
Collapse
Affiliation(s)
- Zhiqiang Pei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Jin Qiu
- Department of Cardiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Rui Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Wei Luo
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Xingxing Cai
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 201322, China
| | - Bin Liu
- Department of Cardiology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Han Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Jiasheng Yin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Yizhe Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Chenguang Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Li Shen
- National Clinical Research for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| |
Collapse
|
3
|
Párraga R, Real C, Fernández-Jiménez R. Cardiovascular magnetic resonance in the working diagnosis of MINOCA: the sooner, the better? REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024; 77:524-526. [PMID: 38316355 DOI: 10.1016/j.rec.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/07/2024]
Affiliation(s)
- Rocío Párraga
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Servicio de Cardiología, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Carlos Real
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Servicio de Cardiología, Hospital Universitario Clínico San Carlos, Madrid, Spain. https://twitter.com/@CHI_Lab_CNIC
| | - Rodrigo Fernández-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Servicio de Cardiología, Hospital Universitario Clínico San Carlos, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
4
|
Juncà G, Teis A, Kasa G, Ferrer-Sistach E, Vallejo N, López-Ayerbe J, Cediel G, Bayés-Genís A, Delgado V. Timing of cardiac magnetic resonance and diagnostic yield in patients with myocardial infarction with nonobstructive coronary arteries. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024; 77:515-523. [PMID: 38061424 DOI: 10.1016/j.rec.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION AND OBJECTIVES The present study sought to establish the diagnostic yield of cardiovascular magnetic resonance (CMR) in a large cohort of patients admitted with myocardial infarction (MI) with nonobstructive coronary artery disease (MINOCA) based on the timing of referral to CMR. METHODS Consecutive patients referred to CMR from January 2009 to February 2022 with a working diagnosis of MINOCA were retrospectively evaluated. Cine, T2-weighted, early, and late gadolinium-enhanced images were acquired and analyzed. The frequency of the underlying diagnosis and the association between timing of CMR and relative frequency of each diagnosis were assessed. RESULTS We included 207 patients (median age 50 years, 60% men). Final diagnosis after CMR was achieved in 91% of the patients (myocarditis in 45%, MI in 20%, tako-tsubo cardiomyopathy in 19%, and other cardiomyopathies in 7%). The performance of CMR within 7 days of admission with MINOCA (median, 5 days; 117 patients) allowed a higher diagnostic yield compared with CMR performed later (median, 10 days; 88 patients) (96% vs 86%, P=.02). Although myocarditis was the most frequent diagnosis in both groups according to time to CMR, its frequency was higher among patients with a CMR performed within the first 7 days (53% vs 35%, P=.02). The frequency of other underlying diagnoses was not influenced by CMR timing. CONCLUSIONS CMR led to an underlying diagnosis of MINOCA in 91% of patients and its diagnostic yield increased to 96% when CMR was performed within 7 days of admission. The most frequent diagnosis was myocarditis..
Collapse
Affiliation(s)
- Gladys Juncà
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Albert Teis
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Gizem Kasa
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Elena Ferrer-Sistach
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Nuria Vallejo
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Jorge López-Ayerbe
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Germán Cediel
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain; Departamento de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Antoni Bayés-Genís
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain; Departamento de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Victoria Delgado
- Instituto del Corazón, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.
| |
Collapse
|
5
|
Welt FGP, Batchelor W, Spears JR, Penna C, Pagliaro P, Ibanez B, Drakos SG, Dangas G, Kapur NK. Reperfusion Injury in Patients With Acute Myocardial Infarction: JACC Scientific Statement. J Am Coll Cardiol 2024; 83:2196-2213. [PMID: 38811097 DOI: 10.1016/j.jacc.2024.02.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 05/31/2024]
Abstract
Despite impressive improvements in the care of patients with ST-segment elevation myocardial infarction, mortality remains high. Reperfusion is necessary for myocardial salvage, but the abrupt return of flow sets off a cascade of injurious processes that can lead to further necrosis. This has been termed myocardial ischemia-reperfusion injury and is the subject of this review. The pathologic and molecular bases for myocardial ischemia-reperfusion injury are increasingly understood and include injury from reactive oxygen species, inflammation, calcium overload, endothelial dysfunction, and impaired microvascular flow. A variety of pharmacologic strategies have been developed that have worked well in preclinical models and some have shown promise in the clinical setting. In addition, there are newer mechanical approaches including mechanical unloading of the heart prior to reperfusion that are in current clinical trials.
Collapse
Affiliation(s)
- Frederick G P Welt
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA.
| | | | - J Richard Spears
- Department of Cardiovascular Medicine, Beaumont Systems, Royal Oak, Michigan, USA
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Department of Cardiology, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Stavros G Drakos
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - George Dangas
- Division of Cardiology, Mount Sinai Health System, New York, New York, USA
| | - Navin K Kapur
- The CardioVascular Center and Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Gissler MC, Antiochos P, Ge Y, Heydari B, Gräni C, Kwong RY. Cardiac Magnetic Resonance Evaluation of LV Remodeling Post-Myocardial Infarction: Prognosis, Monitoring and Trial Endpoints. JACC Cardiovasc Imaging 2024:S1936-878X(24)00127-X. [PMID: 38819335 DOI: 10.1016/j.jcmg.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 06/01/2024]
Abstract
Adverse left ventricular remodeling (ALVR) and subsequent heart failure after myocardial infarction (MI) remain a major cause of patient morbidity and mortality worldwide. Overt inflammation has been identified as the common pathway underlying myocardial fibrosis and development of ALVR post-MI. With its ability to simultaneously provide information about cardiac structure, function, perfusion, and tissue characteristics, cardiac magnetic resonance (CMR) is well poised to inform prognosis and guide early surveillance and therapeutics in high-risk cohorts. Further, established and evolving CMR-derived biomarkers may serve as clinical endpoints in prospective trials evaluating the efficacy of novel anti-inflammatory and antifibrotic therapies. This review provides an overview of post-MI ALVR and illustrates how CMR may help clinical adoption of novel therapies via mechanistic or prognostic imaging markers.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Panagiotis Antiochos
- Cardiology and Cardiac MR Centre, University Hospital Lausanne, Lausanne, Switzerland
| | - Yin Ge
- Division of Cardiology, St Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Bobak Heydari
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christoph Gräni
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raymond Y Kwong
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
7
|
Dawkins S, Digby JE, Belgard TG, Lee R, De Maria GL, Banning AP, Kharbanda RK, Mayr M, Choudhury RP, Channon KM. Stratification of acute myocardial and endothelial cell injury, salvage index and final infarct size by systematic microRNA profiling in acute ST-elevation myocardial infarction. Coron Artery Dis 2024; 35:122-134. [PMID: 38009375 DOI: 10.1097/mca.0000000000001284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
AIM Acute injury and subsequent remodelling responses to ST-segment elevation myocardial infarction (STEMI) are major determinants of clinical outcome. Current imaging and plasma biomarkers provide delayed readouts of myocardial injury and recovery. Here, we sought to systematically characterize all microRNAs (miRs) released during the acute phase of STEMI and relate miR release to magnetic resonance imaging (MRI) findings to predict acute and late responses to STEMI, from a single early blood sample. METHODS AND RESULTS miRs were quantified in blood samples obtained from patients after primary PCI (PPCI) for STEMI. Cardiac MRI (cMRI) was performed to quantify myocardial edema, infarct size and salvage index. Regression models were constructed to predict these outcomes measures, which were then tested with a validation cohort. Transcoronary miR release was quantified from paired measurements of coronary artery and coronary sinus samples. A cell culture model was used to identify endothelial cell-derived miRs.A total of 72 patients undergoing PPCI for acute STEMI underwent miR analysis and cMRI. About >200 miRs were detectable in plasma after STEMI, from which 128 miRs were selected for quantification in all patients. Known myocardial miRs demonstrated a linear correlation with troponin release, and these increased across the transcoronary gradient. We identified novel miRs associated with microvascular injury and myocardial salvage. Regression models were constructed using a training cohort, then tested in a validation cohort, and predicted myocardial oedema, infarct size and salvage index. CONCLUSION Analysis of miR release after STEMI identifies biomarkers that predict both acute and late outcomes after STEMI. A novel miR-based biomarker score enables the estimation of area at risk, late infarct size and salvage index from a single blood sample 6 hours after PPCI, providing a simple and rapid alternative to serial cMRI characterization of STEMI outcome.
Collapse
Affiliation(s)
- Sam Dawkins
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Janet E Digby
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | - Regent Lee
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Giovanni Luigi De Maria
- Oxford Heart Centre, National Institute for Health (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford
| | - Adrian P Banning
- Oxford Heart Centre, National Institute for Health (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford
| | - Rajesh K Kharbanda
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Oxford Heart Centre, National Institute for Health (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Oxford Heart Centre, National Institute for Health (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Oxford Heart Centre, National Institute for Health (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford
| |
Collapse
|
8
|
Binek A, Castans C, Jorge I, Bagwan N, Rodríguez JM, Fernández-Jiménez R, Galán-Arriola C, Oliver E, Gómez M, Clemente-Moragón A, Ibanez B, Camafeita E, Vázquez J. Oxidative Post-translational Protein Modifications upon Ischemia/Reperfusion Injury. Antioxidants (Basel) 2024; 13:106. [PMID: 38247530 PMCID: PMC10812827 DOI: 10.3390/antiox13010106] [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: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
While reperfusion, or restoration of coronary blood flow in acute myocardial infarction, is a requisite for myocardial salvage, it can paradoxically induce a specific damage known as ischemia/reperfusion (I/R) injury. Our understanding of the precise pathophysiological molecular alterations leading to I/R remains limited. In this study, we conducted a comprehensive and unbiased time-course analysis of post-translational modifications (PTMs) in the post-reperfused myocardium of two different animal models (pig and mouse) and evaluated the effect of two different cardioprotective therapies (ischemic preconditioning and neutrophil depletion). In pigs, a first wave of irreversible oxidative damage was observed at the earliest reperfusion time (20 min), impacting proteins essential for cardiac contraction. A second wave, characterized by irreversible oxidation on different residues and reversible Cys oxidation, occurred at late stages (6-12 h), affecting mitochondrial, sarcomere, and inflammation-related proteins. Ischemic preconditioning mitigated the I/R damage caused by the late oxidative wave. In the mouse model, the two-phase pattern of oxidative damage was replicated, and neutrophil depletion mitigated the late wave of I/R-related damage by preventing both Cys reversible oxidation and irreversible oxidation. Altogether, these data identify protein PTMs occurring late after reperfusion as an actionable therapeutic target to reduce the impact of I/R injury.
Collapse
Grants
- PGC2018-097019-B-I00, PID2021-122348NB-I00, PID2022-140176OB-I00 Spanish Ministry of Science, Innovation and Universities
- Fondo de Investigación Sanitaria grant PRB3 PT17/0019/0003- ISCIII-SGEFI / ERDF, ProteoRed Instituto de Salud Carlos III
- IMMUNO-VAR, P2022/BMD-7333, and RENIM-CM, P2022/BMD-7403 Comunidad de Madrid
- HR17-00247, HR22-00533 and HR22-00253 "la Caixa" Banking Foundation
- ERC Consolidator Grant "MATRIX", 819775 European Commission
- grant PI22/01560 ISCIII-Fondo de Investigación Sanitaria and European Union
- FP7-PEOPLE-2013-ITN-Cardionext European Union's Seventh Framework Programme
- Formacion del Profesorado Universitario (FPU14/05292) Spanish Ministry of Education, Culture and Sports
- PID2021-133167OB-100, RYC2020-028884-I, CEX2020-001041-S MCIN/AEI/10.13039/501100011033
Collapse
Affiliation(s)
- Aleksandra Binek
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Celia Castans
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Inmaculada Jorge
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Navratan Bagwan
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - José Manuel Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Rodrigo Fernández-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Department of Cardiology, Hospital Universitario Clínico San Carlos, Profesor Martín Lagos, s/n, 28040 Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Mónica Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- IIS-Fundación Jiménez Díaz Hospital, Avenida Reyes Católicos, 2, 28040 Madrid, Spain
| | - Emilio Camafeita
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
9
|
Reiss S, Thielmann J, Fischer J, Lottner T, Maier A, Westermann D, von Zur Mühlen C, Heidt T, Bock M. Combination of high resolution MRI with 3D-printed needle guides for ex vivo myocardial biopsies. Sci Rep 2024; 14:606. [PMID: 38182761 PMCID: PMC10770147 DOI: 10.1038/s41598-023-50943-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024] Open
Abstract
Magnetic resonance imaging (MRI) provides a multitude of techniques to detect and characterize myocardial infarction. To correlate MRI findings with histology, in most cases terminal animal studies are performed; however, precise extraction and spatial correlation of myocardial tissue samples to MRI image data is difficult. In this proof of concept study, we present a 3D-printing technique to facilitate the extraction of tissue samples from myocardial regions. Initially, seven pig hearts embedded in formaldehyde were imaged on a clinical 3 T system to define biopsy targets on high resolution ex vivo images. Magnitude images and R2*-maps acquired with a 3D multi-echo gradient echo sequence and 0.58 mm isotropic resolution were used to create digital models of the cardiac anatomy. Biopsy guides were 3D-printed to steer the extraction of myocardial samples. In total, 61 tissue samples were extracted with an average offset of the tissue sample location from the target location of 0.59 ± 0.36 mm. This offset was not dependent on the distance of the target point to the epicardial surface. Myocardial tissue could be extracted from all samples. The presented method enables extraction of myocardial tissue samples that are selected by ex vivo MRI with submillimeter precision.
Collapse
Affiliation(s)
- Simon Reiss
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany.
| | - Julien Thielmann
- Department of Cardiology and Angiology, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Johannes Fischer
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany
| | - Thomas Lottner
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany
| | - Alexander Maier
- Department of Cardiology and Angiology, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Constantin von Zur Mühlen
- Department of Cardiology and Angiology, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Timo Heidt
- Department of Cardiology and Angiology, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Michael Bock
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Medical Center Freiburg, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany
| |
Collapse
|
10
|
Ndrepepa G, Kastrati A. Coronary No-Reflow after Primary Percutaneous Coronary Intervention-Current Knowledge on Pathophysiology, Diagnosis, Clinical Impact and Therapy. J Clin Med 2023; 12:5592. [PMID: 37685660 PMCID: PMC10488607 DOI: 10.3390/jcm12175592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Coronary no-reflow (CNR) is a frequent phenomenon that develops in patients with ST-segment elevation myocardial infarction (STEMI) following reperfusion therapy. CNR is highly dynamic, develops gradually (over hours) and persists for days to weeks after reperfusion. Microvascular obstruction (MVO) developing as a consequence of myocardial ischemia, distal embolization and reperfusion-related injury is the main pathophysiological mechanism of CNR. The frequency of CNR or MVO after primary PCI differs widely depending on the sensitivity of the tools used for diagnosis and timing of examination. Coronary angiography is readily available and most convenient to diagnose CNR but it is highly conservative and underestimates the true frequency of CNR. Cardiac magnetic resonance (CMR) imaging is the most sensitive method to diagnose MVO and CNR that provides information on the presence, localization and extent of MVO. CMR imaging detects intramyocardial hemorrhage and accurately estimates the infarct size. MVO and CNR markedly negate the benefits of reperfusion therapy and contribute to poor clinical outcomes including adverse remodeling of left ventricle, worsening or new congestive heart failure and reduced survival. Despite extensive research and the use of therapies that target almost all known pathophysiological mechanisms of CNR, no therapy has been found that prevents or reverses CNR and provides consistent clinical benefit in patients with STEMI undergoing reperfusion. Currently, the prevention or alleviation of MVO and CNR remain unmet goals in the therapy of STEMI that continue to be under intense research.
Collapse
Affiliation(s)
- Gjin Ndrepepa
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany;
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstrasse 36, 80636 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
11
|
Alkhalil M, De Maria GL, Akbar N, Ruparelia N, Choudhury RP. Prospects for Precision Medicine in Acute Myocardial Infarction: Patient-Level Insights into Myocardial Injury and Repair. J Clin Med 2023; 12:4668. [PMID: 37510783 PMCID: PMC10380764 DOI: 10.3390/jcm12144668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The past decade has seen a marked expansion in the understanding of the pathobiology of acute myocardial infarction and the systemic inflammatory response that it elicits. At the same time, a portfolio of tools has emerged to characterise some of these processes in vivo. However, in clinical practice, key decision making still largely relies on assessment built around the timing of the onset of chest pain, features on electrocardiograms and measurements of plasma troponin. Better understanding the heterogeneity of myocardial injury and patient-level responses should provide new opportunities for diagnostic stratification to enable the delivery of more rational therapies. Characterisation of the myocardium using emerging imaging techniques such as the T1, T2 and T2* mapping techniques can provide enhanced assessments of myocardial statuses. Physiological measures, which include microcirculatory resistance and coronary flow reserve, have been shown to predict outcomes in AMI and can be used to inform treatment selection. Functionally informative blood biomarkers, including cellular transcriptomics; microRNAs; extracellular vesicle analyses and soluble markers, all give insights into the nature and timing of the innate immune response and its regulation in acute MI. The integration of these and other emerging tools will be key to developing a fuller understanding of the patient-level processes of myocardial injury and repair and should fuel new possibilities for rational therapeutic intervention.
Collapse
Affiliation(s)
- Mohammad Alkhalil
- Cardiothoracic Centre, Freeman Hospital, Newcastle-upon-Tyne NE7 7DN, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK
| | | | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Neil Ruparelia
- Cardiology Department, Hammersmith Hospital, Imperial College London, London W12 0HS, UK
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
12
|
Mileva N, Bergamaschi L, Paolisso P, Pizzi C, Vassilev D. Reply: MINOCA and CMR: Where Do We Stand? JACC Cardiovasc Imaging 2023; 16:997. [PMID: 37407130 DOI: 10.1016/j.jcmg.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/10/2023] [Indexed: 07/07/2023]
|
13
|
Kleinbongard P, Lieder HR, Skyschally A, Heusch G. No robust reduction of infarct size and no-reflow by metoprolol pretreatment in adult Göttingen minipigs. Basic Res Cardiol 2023; 118:23. [PMID: 37289247 PMCID: PMC10250284 DOI: 10.1007/s00395-023-00993-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/09/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023]
Abstract
Whereas prior experiments in juvenile pigs had reported infarct size reduction by intravenous metoprolol early during myocardial ischaemia, two major clinical trials in patients with reperfused acute myocardial infarction were equivocal. We, therefore, went back and tested the translational robustness of infarct size reduction by metoprolol in minipigs. Using a power analysis-based prospective design, we pretreated 20 anaesthetised adult Göttingen minipigs with 1 mg kg-1 metoprolol or placebo and subjected them to 60-min coronary occlusion and 180-min reperfusion. Primary endpoint was infarct size (triphenyl tetrazolium chloride staining) as a fraction of area at risk; no-reflow area (thioflavin-S staining) was a secondary endpoint. There was no significant reduction in infarct size (46 ± 8% of area at risk with metoprolol vs. 42 ± 8% with placebo) or area of no-reflow (19 ± 21% of infarct size with metoprolol vs. 15 ± 23% with placebo). However, the inverse relationship between infarct size and ischaemic regional myocardial blood flow was modestly, but significantly shifted downwards with metoprolol, whereas ischaemic blood flow tended to be reduced by metoprolol. With an additional dose of 1 mg kg-1 metoprolol after 30-min ischaemia in 4 additional pigs, infarct size was also not reduced (54 ± 9% vs. 46 ± 8% in 3 contemporary placebo, n.s.), and area of no-reflow tended to be increased (59 ± 20% vs. 29 ± 12%, n.s.).Infarct size reduction by metoprolol in pigs is not robust, and this result reflects the equivocal clinical trials. The lack of infarct size reduction may be the result of opposite effects of reduced infarct size at any given blood flow and reduced blood flow, possibly through unopposed alpha-adrenergic coronary vasoconstriction.
Collapse
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Helmut Raphael Lieder
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
14
|
Blaszczyk E, Hellwig S, Saad H, Ganeshan R, Stengl H, Nolte CH, Fiebach JB, Endres M, Kuhnt J, Gröschel J, Schulz-Menger J, Scheitz JF. Myocardial injury in patients with acute ischemic stroke detected by cardiovascular magnetic resonance imaging. Eur J Radiol 2023; 165:110908. [PMID: 37315403 DOI: 10.1016/j.ejrad.2023.110908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/29/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Patients with acute ischemic stroke (AIS) are at high risk of adverse cardiovascular events. Until now, the burden of myocardial injury derived from cardiovascular magnetic resonance imaging (CMR) has not been established in this population. METHODS Patients with AIS underwent CMR at 3 Tesla within 120 h after the index stroke as part of a prospective, single-center study. Patients with persistent atrial fibrillation were excluded. Morphology and function of both cardiac chambers and atria were assessed applying SSFP cine. Myocardial tissue differentiation was based on native and contrast-enhanced imaging including late gadolinium enhancement (LGE) after 0.15 mmol/kg gadobutrol for focal fibrosis and parametric T2- and T1-mapping for diffuse findings. To detect myocardial deformation global longitudinal (GLS), circumferential (GCS) and radial (GRS) strain was measured applying feature tracking. Cardiac troponin was measured using a high-sensitivity assay (99th percentile upper reference limit 14 ng/L). T2 mapping values were compared with 20 healthy volunteers. RESULTS CMR with contrast media was successfully performed in 92 of 115 patients (mean age 74 years, 40% female, known myocardial infarction 6%). Focal myocardial fibrosis (LGE) was detected in 31 of 92 patients (34%) of whom 23/31 (74%) showed an ischemic pattern. Patients with LGE were more likely to have diabetes, prior myocardial infarction, prior ischemic stroke, and to have elevated troponin levels compared to those without. Presence of LGE was accompanied by diffuse fibrosis (increased T1 native values) even in remote cardiac areas as well as reduced global radial, circumferential and longitudinal strain values. In 14/31 (45%) of all patients with LGE increased T2-mapping values were detectable. CONCLUSIONS More than one-third of patients with AIS have evidence of focal myocardial fibrosis on CMR. Nearly half of these changes may have acute or subacute onset. These findings are accompanied by diffuse myocardial changes and reduced myocardial deformation. Further studies, ideally with serial CMR measurements during follow-up, are required to establish the impact of these findings on long-term prognosis after AIS.
Collapse
Affiliation(s)
- E Blaszczyk
- Charité Universitätsmedizin Berlin, Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine , HELIOS Klinikum Berlin Buch, Cardiology, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - S Hellwig
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany
| | - H Saad
- Charité Universitätsmedizin Berlin, Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine , HELIOS Klinikum Berlin Buch, Cardiology, Berlin, Germany
| | - R Ganeshan
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany
| | - H Stengl
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany
| | - C H Nolte
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Germany
| | - J B Fiebach
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany
| | - M Endres
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany; ExcellenceCluster NeuroCure, Germany; German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Germany; Berlin Institute of Health (BIH), Germany
| | - J Kuhnt
- Charité Universitätsmedizin Berlin, Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine , HELIOS Klinikum Berlin Buch, Cardiology, Berlin, Germany
| | - J Gröschel
- Charité Universitätsmedizin Berlin, Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine , HELIOS Klinikum Berlin Buch, Cardiology, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - J Schulz-Menger
- Charité Universitätsmedizin Berlin, Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine , HELIOS Klinikum Berlin Buch, Cardiology, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - J F Scheitz
- Klinik für Neurologie mit Experimenteller Neurologie, Charité Universitätsmedizin Berlin und Centrum für Schlaganfallforschung, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
15
|
Consegal M, Barba I, García Del Blanco B, Otaegui I, Rodríguez-Palomares JF, Martí G, Serra B, Bellera N, Ojeda-Ramos M, Valente F, Carmona MÁ, Miró-Casas E, Sambola A, Lidón RM, Bañeras J, Barrabés JA, Rodríguez C, Benito B, Ruiz-Meana M, Inserte J, Ferreira-González I, Rodríguez-Sinovas A. Spontaneous reperfusion enhances succinate concentration in peripheral blood from stemi patients but its levels does not correlate with myocardial infarct size or area at risk. Sci Rep 2023; 13:6907. [PMID: 37106099 PMCID: PMC10140265 DOI: 10.1038/s41598-023-34196-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 04/25/2023] [Indexed: 04/29/2023] Open
Abstract
Succinate is enhanced during initial reperfusion in blood from the coronary sinus in ST-segment elevation myocardial infarction (STEMI) patients and in pigs submitted to transient coronary occlusion. Succinate levels might have a prognostic value, as they may correlate with edema volume or myocardial infarct size. However, blood from the coronary sinus is not routinely obtained in the CathLab. As succinate might be also increased in peripheral blood, we aimed to investigate whether peripheral plasma concentrations of succinate and other metabolites obtained during coronary revascularization correlate with edema volume or infarct size in STEMI patients. Plasma samples were obtained from peripheral blood within the first 10 min of revascularization in 102 STEMI patients included in the COMBAT-MI trial (initial TIMI 1) and from 9 additional patients with restituted coronary blood flow (TIMI 2). Metabolite concentrations were analyzed by 1H-NMR. Succinate concentration averaged 0.069 ± 0.0073 mmol/L in patients with TIMI flow ≤ 1 and was significantly increased in those with TIMI 2 at admission (0.141 ± 0.058 mmol/L, p < 0.05). However, regression analysis did not detect any significant correlation between most metabolite concentrations and infarct size, extent of edema or other cardiac magnetic resonance (CMR) variables. In conclusion, spontaneous reperfusion in TIMI 2 patients associates with enhanced succinate levels in peripheral blood, suggesting that succinate release increases overtime following reperfusion. However, early plasma levels of succinate and other metabolites obtained from peripheral blood does not correlate with the degree of irreversible injury or area at risk in STEMI patients, and cannot be considered as predictors of CMR variables.Trial registration: Registered at www.clinicaltrials.gov (NCT02404376) on 31/03/2015. EudraCT number: 2015-001000-58.
Collapse
Affiliation(s)
- Marta Consegal
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ignasi Barba
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Medicine, University of Vic - Central University of Catalonia (UVicUCC), Can Baumann. Ctra. de Roda, 70, 08500, Vic, Spain
| | - Bruno García Del Blanco
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Imanol Otaegui
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - José F Rodríguez-Palomares
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Gerard Martí
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Bernat Serra
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Neus Bellera
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Ojeda-Ramos
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Filipa Valente
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Ángeles Carmona
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisabet Miró-Casas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonia Sambola
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa María Lidón
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Bañeras
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - José Antonio Barrabés
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Rodríguez
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Begoña Benito
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Ferreira-González
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red (CIBER) de Epidemiología y Salud Pública, CIBERESP, Instituto de Salud Carlos III, Madrid, Spain.
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
16
|
Urzua Fresno C, Sanchez Tijmes F, Shaw KE, Huang F, Thavendiranathan P, Khullar S, Seidman MA, Hanneman K. Cardiac Imaging in Myocarditis: Current Evidence and Future Directions. Can Assoc Radiol J 2023; 74:147-159. [PMID: 36062360 DOI: 10.1177/08465371221119713] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Myocarditis is defined as a non-ischemic inflammatory disease of the myocardium. It remains a challenge to diagnose given non-specific symptoms and lack of specific blood biomarkers. Cardiac imaging plays an important role in the evaluation of myocarditis with unique strengths and limitations of different imaging modalities, including cardiac magnetic resonance imaging, echocardiography, cardiac computed tomography, and positron emission tomography. The purpose of this review is to discuss the strengths and limitations of various cardiac imaging techniques in the evaluation of myocarditis, review imaging findings in specific causes of myocarditis including COVID-19 and after vaccination, evaluate the role of imaging in differentiating myocarditis from potential mimics and differential considerations, identify current gaps in knowledge, and propose future directions.
Collapse
Affiliation(s)
- Camila Urzua Fresno
- Department of Medical Imaging, Toronto General Hospital, Peter Munk Cardiac Center, University Health Network (UHN), 7938University of Toronto, Toronto, ON, Canada
| | - Felipe Sanchez Tijmes
- Department of Medical Imaging, Toronto General Hospital, Peter Munk Cardiac Center, University Health Network (UHN), 7938University of Toronto, Toronto, ON, Canada.,Department of Medical Imaging, Clinica Santa Maria, 33179Universidad de los Andes, Santiago, Chile
| | - Kirsten E Shaw
- Department of Cardiology, 123769Hennepin Healthcare/Minneapolis Heart Institute, Minneapolis, MN, USA
| | - Flora Huang
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Paaladinesh Thavendiranathan
- Department of Medical Imaging, Toronto General Hospital, Peter Munk Cardiac Center, University Health Network (UHN), 7938University of Toronto, Toronto, ON, Canada.,Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network (UHN), 7938University of Toronto, Toronto, ON, Canada
| | - Sharmila Khullar
- Department of Laboratory Medicine & Pathobiology, 7938University of Toronto, Toronto, ON, Canada
| | - Michael A Seidman
- Department of Laboratory Medicine & Pathobiology, 7938University of Toronto, Toronto, ON, Canada.,Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - Kate Hanneman
- Department of Medical Imaging, Toronto General Hospital, Peter Munk Cardiac Center, University Health Network (UHN), 7938University of Toronto, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network (UHN), 7938University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Radike M, Sutelman P, Ben-Aicha S, Gutiérrez M, Mendieta G, Alcover S, Casaní L, Arderiu G, Borrell-Pages M, Padró T, Badimon L, Vilahur G. A comprehensive and longitudinal cardiac magnetic resonance imaging study of the impact of coronary ischemia duration on myocardial damage in a highly translatable animal model. Eur J Clin Invest 2023; 53:e13860. [PMID: 35986736 DOI: 10.1111/eci.13860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/01/2022] [Accepted: 08/18/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES We performed a comprehensive assessment of the effect of myocardial ischemia duration on cardiac structural and functional parameters by serial cardiac magnetic resonance (CMR) and characterized the evolving scar. BACKGROUND CMR follow-up on the cardiac impact of time of ischemia in a closed-chest animal model of myocardial infarction with human resemblance is missing. METHODS Pigs underwent MI induction by occlusion of the left anterior descending (LAD) coronary artery for 30, 60, 90 or 120 min and then revascularized. Serial CMR was performed on day 3 and day 42 post-MI. CMR measurements were also run in a sham-operated group. Cellular and molecular changes were investigated. RESULTS On day 3, cardiac damage and function were similar in sham and pigs subjected to 30 min of ischemia. Cardiac damage (oedema and necrosis) significantly increased from 60 min onwards. Microvascular obstruction was extensively seen in animals with ≥90 min of ischemia and correlated with cardiac damage. A drop in global systolic function and wall motion of the jeopardized segments was seen in pigs subjected to ≥60 min of ischemia. On day 42, scar size and cardiac dysfunction followed the same pattern in the animals subjected to ≥60 min of ischemia. Adverse left ventricular remodelling (worsening of both LV volumes) was only present in animals subjected to 120 min of ischemia. Cardiac fibrosis, myocyte hypertrophy and vessel rarefaction were similar in the infarcted myocardium of pigs subjected to ≥60 min of ischemia. No changes were observed in the remote myocardium. CONCLUSION Sixty-minute LAD coronary occlusion already induces cardiac structural and functional alterations with longer ischemic time (120 min) causing adverse LV remodelling.
Collapse
Affiliation(s)
- Monika Radike
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Radiology Department, Liverpool Heart and Chest Hospital NHS Foundation Trust, Liverpool, UK
| | - Pablo Sutelman
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Soumaya Ben-Aicha
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Manuel Gutiérrez
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Radiology Department, Liverpool Heart and Chest Hospital NHS Foundation Trust, Liverpool, UK
| | - Guiomar Mendieta
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sebastià Alcover
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Laura Casaní
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Gemma Arderiu
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - María Borrell-Pages
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain.,Cardiovascular Research Chair UAB, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Shanmuganathan M, Masi A, Burrage MK, Kotronias RA, Borlotti A, Scarsini R, Banerjee A, Terentes-Printzios D, Zhang Q, Hann E, Tunnicliffe E, Lucking A, Langrish J, Kharbanda R, De Maria GL, Banning AP, Choudhury RP, Channon KM, Piechnik SK, Ferreira VM. Acute Response in the Noninfarcted Myocardium Predicts Long-Term Major Adverse Cardiac Events After STEMI. JACC Cardiovasc Imaging 2023; 16:46-59. [PMID: 36599569 PMCID: PMC9834063 DOI: 10.1016/j.jcmg.2022.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Acute ST-segment elevation myocardial infarction (STEMI) has effects on the myocardium beyond the immediate infarcted territory. However, pathophysiologic changes in the noninfarcted myocardium and their prognostic implications remain unclear. OBJECTIVES The purpose of this study was to evaluate the long-term prognostic value of acute changes in both infarcted and noninfarcted myocardium post-STEMI. METHODS Patients with acute STEMI undergoing primary percutaneous coronary intervention underwent evaluation with blood biomarkers and cardiac magnetic resonance (CMR) at 2 days and 6 months, with long-term follow-up for major adverse cardiac events (MACE). A comprehensive CMR protocol included cine, T2-weighted, T2∗, T1-mapping, and late gadolinium enhancement (LGE) imaging. Areas without LGE were defined as noninfarcted myocardium. MACE was a composite of cardiac death, sustained ventricular arrhythmia, and new-onset heart failure. RESULTS Twenty-two of 219 patients (10%) experienced an MACE at a median of 4 years (IQR: 2.5-6.0 years); 152 patients returned for the 6-month visit. High T1 (>1250 ms) in the noninfarcted myocardium was associated with lower left ventricular ejection fraction (LVEF) (51% ± 8% vs 55% ± 9%; P = 0.002) and higher NT-pro-BNP levels (290 pg/L [IQR: 103-523 pg/L] vs 170 pg/L [IQR: 61-312 pg/L]; P = 0.008) at 6 months and a 2.5-fold (IQR: 1.03-6.20) increased risk of MACE (2.53 [IQR: 1.03-6.22]), compared with patients with normal T1 in the noninfarcted myocardium (P = 0.042). A lower T1 (<1,300 ms) in the infarcted myocardium was associated with increased MACE (3.11 [IQR: 1.19-8.13]; P = 0.020). Both noninfarct and infarct T1 were independent predictors of MACE (both P = 0.001) and significantly improved risk prediction beyond LVEF, infarct size, and microvascular obstruction (C-statistic: 0.67 ± 0.07 vs 0.76 ± 0.06, net-reclassification index: 40% [IQR: 12%-64%]; P = 0.007). CONCLUSIONS The acute responses post-STEMI in both infarcted and noninfarcted myocardium are independent incremental predictors of long-term MACE. These insights may provide new opportunities for treatment and risk stratification in STEMI.
Collapse
Affiliation(s)
- Mayooran Shanmuganathan
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ambra Masi
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Matthew K. Burrage
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rafail A. Kotronias
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alessandra Borlotti
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Roberto Scarsini
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Abhirup Banerjee
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Dimitrios Terentes-Printzios
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Qiang Zhang
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Evan Hann
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Elizabeth Tunnicliffe
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Andrew Lucking
- Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jeremy Langrish
- Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rajesh Kharbanda
- Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Giovanni Luigi De Maria
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Adrian P. Banning
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robin P. Choudhury
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Keith M. Channon
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom,Address for correspondence: Prof Keith Channon, Level 2–Oxford Heart Centre, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom.
| | - Stefan K. Piechnik
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Vanessa M. Ferreira
- Acute Vascular Imaging Centre (AVIC), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom,Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom,Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | | |
Collapse
|
19
|
Harris NR, Bálint L, Dy DM, Nielsen NR, Méndez HG, Aghajanian A, Caron KM. The ebb and flow of cardiac lymphatics: a tidal wave of new discoveries. Physiol Rev 2023; 103:391-432. [PMID: 35953269 PMCID: PMC9576179 DOI: 10.1152/physrev.00052.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022] Open
Abstract
The heart is imbued with a vast lymphatic network that is responsible for fluid homeostasis and immune cell trafficking. Disturbances in the forces that regulate microvascular fluid movement can result in myocardial edema, which has profibrotic and proinflammatory consequences and contributes to cardiovascular dysfunction. This review explores the complex relationship between cardiac lymphatics, myocardial edema, and cardiac disease. It covers the revised paradigm of microvascular forces and fluid movement around the capillary as well as the arsenal of preclinical tools and animal models used to model myocardial edema and cardiac disease. Clinical studies of myocardial edema and their prognostic significance are examined in parallel to the recent elegant animal studies discerning the pathophysiological role and therapeutic potential of cardiac lymphatics in different cardiovascular disease models. This review highlights the outstanding questions of interest to both basic scientists and clinicians regarding the roles of cardiac lymphatics in health and disease.
Collapse
Affiliation(s)
- Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - László Bálint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Danielle M Dy
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hernán G Méndez
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Amir Aghajanian
- Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
20
|
Hashimoto Y, Soeda T, Seno A, Okayama S, Fukuda N, Yano H, Iwai A, Nogi K, Hirai K, Fujimoto H, Suzuki M, Iwama H, Nakai T, Doi N, Saito Y. Reverse Remodeling and Non-Contrast T1 Hypointense Infarct Core in Patients With Reperfused Acute Myocardial Infarction. Circ J 2022; 86:1968-1979. [PMID: 36288957 DOI: 10.1253/circj.cj-22-0479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
BACKGROUND Non-contrast T1 hypointense infarct cores (ICs) within infarcted myocardium detected using cardiac magnetic resonance imaging (CMR) T1 mapping may help assess the severity of left ventricular (LV) injury. However, because the relationship of ICs with chronic LV reverse remodeling (LVRR) is unknown, this study aimed to clarify it. METHODS AND RESULTS We enrolled patients with reperfused AMI who underwent baseline CMR on day-7 post-primary percutaneous coronary intervention (n=109) and 12-month follow-up CMR (n=94). Correlations between ICs and chronic LVRR (end-systolic volume decrease ≥15% at 12-month follow-up from baseline CMR) were investigated. We detected 52 (47.7%) ICs on baseline CMR by non-contrast-T1 mapping. LVRR was found in 52.1% of patients with reperfused AMI at 12-month follow-up. Patients with ICs demonstrated higher peak creatine kinase levels, higher B-type natriuretic peptide levels at discharge, lower LV ejection fraction at discharge, and lower incidence of LVRR than those without ICs (26.5% vs. 73.3%, P<0.001) at follow-up. Multivariate logistic regression analysis showed that the presence of ICs was an independent and the strongest negative predictor for LVRR at 12-month follow-up (hazard ratio: 0.087, 95% confidence interval: 0.017-0.459, P=0.004). Peak creatine kinase levels, native T1 values at myocardial edema, and myocardial salvaged indices also correlated with ICs. CONCLUSIONS ICs detected by non-contrast-T1 mapping with 3.0-T CMR were an independent negative predictor of LVRR in patients with reperfused AMI.
Collapse
Affiliation(s)
| | | | - Ayako Seno
- Cardiovascular Medicine, Nara Medical University
| | - Satoshi Okayama
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Nozomi Fukuda
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Hiroki Yano
- Cardiovascular Medicine, Nara Medical University
| | - Atsushi Iwai
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | | | - Kaeko Hirai
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Hajime Fujimoto
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Megumi Suzuki
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Hajime Iwama
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Takehito Nakai
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Naofumi Doi
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| | - Yoshihiko Saito
- Cardiovascular Medicine, Nara Medical University
- Cardiovascular Medicine, Nara Prefecture Seiwa Medical Center
| |
Collapse
|
21
|
The role of temporal changes of pro-inflammatory cytokines in the development of adverse cardiac remodeling after ST-elevation myocardial infarction. Adv Cardiol 2022; 18:217-227. [PMID: 36751290 PMCID: PMC9885240 DOI: 10.5114/aic.2022.120938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022]
Abstract
Introduction Increasing evidence supports the view that pro-inflammatory cytokines play a role in fibrosis after myocardial infarction (MI). It has been suggested that interleukin (IL)-12p40, a pro-inflammatory cytokine, can induce interferon γ (IFN-γ) and matrix metalloproteinase (MMP). However, the role of IL-12p40 in adverse cardiac remodeling (AR) after ST-elevation MI (STEMI) is unclear. Aim To examine the role of temporal changes of pro-inflammatory cytokines in the development of post-STEMI AR. Material and methods A total of 43 patients with STEMI for the first time ever were prospectively analyzed. In cardiac magnetic resonance imaging at 6 months after STEMI, a decrease of left ventricular end-diastolic volume by ≥ 12% was defined as reverse cardiac remodeling (RR), and a 12% increase was defined as AR. Cytokine concentrations were measured on the first day (baseline) and 2 weeks after STEMI. Results Mean IL-12p40 (59.1 ±14.5 vs. 46.7 ±9.1 pq/ml, p = 0.001), median IFN-γ (20.4 vs. 16.2 pq/ml, p = 0.048) and median MMP-2 (33866 vs. 20691 pq/ml, p = 0.011) baseline concentrations were higher in AR than RR. In patients with AR, IL-12p40 level was lower at 2 weeks than baseline (p < 0.001). There was a positive correlation between the baseline concentrations of IL-12p40, IFN-γ, MMP-2, C-reactive protein and infarct size (p < 0.05). Increased IL-12p40 and MMP-2 baseline levels were independently associated with AR (OR = 1.14, p = 0.010; OR = 1.08, p = 0.035). Conclusions In the initial phase of MI, greater release of pro-inflammatory cytokines was associated with increased MMP-2 levels. Elevated expression of IL-12 and MMP-2 had an independent association with AR. This may be related to the excessive inflammatory response in the initial phase of MI.
Collapse
|
22
|
Bonfig NL, Soukup CR, Shah AA, Davidson SJ, Stanberry LI, Okeson BK, Traverse JH. Circadian dependence of microvascular obstruction during ST-segment elevation myocardial infarction. Int J Cardiol 2022; 366:25-29. [PMID: 35817202 DOI: 10.1016/j.ijcard.2022.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Microvascular obstruction (MVO) contributes significantly to adverse left-ventricular remodeling and mortality following ST-segment elevation myocardial infarction (STEMI). Because circadian processes contribute significantly to the timing and degree of ischemic injury in STEMI we hypothesized that the occurrence of MVO may also exhibit circadian behavior. METHODS AND RESULTS A single center cohort trial of 336 STEMI patients (273 M 63 F) with their first STEMI who were reperfused with primary percutaneous coronary intervention (PCI) and referred for cardiac MRI prior to discharge. The time of onset of chest pain was recorded from the patients chart and used to stratify patients with MVO over a 24-h cycle to analyze for circadian behavior. Subjects with MVO (n = 200) had greater infarct size by cMRI (45 vs. 20 g; p < 0.001), had reduced ejection fraction (LVEF = 50 vs 45%; p = 0.008) and significantly greater LV end-diastolic (LVEDVI) and end-systolic (LVESVI) volume index compared to subject without MVO (n = 136). The frequency of patients with MVO was compared against the frequency of patients without MVO at each 1-h and 3-h period over a 24-h cycle. A clear peak in patients with MVO (MVO + / MVO -) was seen at the 0700 h interval where 26 out of 27 patients had MVO (p = 0.0038) although MVO mass was not increased. This observation remained significant at the 06-09 time interval when 3-h segments were analyzed. Through 2021, mortality in patients with MVO was significantly greater compared to patients without MVO (n = 20 vs. 5, p < 0.03). CONCLUSIONS This analysis reveals for the first time a circadian dependence of the frequency of MVO in the setting of STEMI which could explain in part, the wide variation in MVO seen in STEMI patients with similar ischemic times and infarct size.
Collapse
Affiliation(s)
- Nicole L Bonfig
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America; The University of Minnesota Medical School, Cardiovascular Division, Minneapolis, MN, United States of America
| | - Chase R Soukup
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America; The University of Minnesota Medical School, Cardiovascular Division, Minneapolis, MN, United States of America
| | - Ananya A Shah
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America
| | - Sarah J Davidson
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America; Duke University School of Medicine, 920 East 28th Street; Suite 300; Minneapolis, MN 55407, United States of America
| | - Larissa I Stanberry
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America
| | - Brynn K Okeson
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America
| | - Jay H Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN, United States of America; The University of Minnesota Medical School, Cardiovascular Division, Minneapolis, MN, United States of America.
| |
Collapse
|
23
|
Targeted trapping of endogenous endothelial progenitor cells for myocardial ischemic injury repair through neutrophil-mediated SPIO nanoparticle-conjugated CD34 antibody delivery and imaging. Acta Biomater 2022; 146:421-433. [PMID: 35545187 DOI: 10.1016/j.actbio.2022.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/22/2022]
Abstract
Endothelia progenitor cell (EPC)-based revascularization therapies have shown promise for the treatment of myocardial ischemic injury. However, applications and efficacy are limited by the relatively inefficient recruitment of endogenous EPCs to the ischemic area, while implantation of exogenous EPCs carries the risk of tumorigenicity. In this study, we developed a therapeutic protocol that relies on the capacity of neutrophils (NEs) to target lesions and release preloaded EPC-binding molecules for high efficiency capture. Neutrophils were loaded with superparamagnetic iron oxide nanoparticles conjugated to an antibody against the EPC surface marker CD34 (SPIO-antiCD34/NEs), and the therapeutic efficacy in ischemic mouse heart following SPIO-antiCD34/NEs injection was monitored by SPIO-enhanced magnetic resonance imaging (MRI). These SPIO-antiCD34/NEs exhibited unimpaired cell viability, superoxide generation, and chemotaxis in vitro as well as satisfactory biocompatibility in vivo. In a mouse model of acute myocardial infarction (MI), SPIO-antiCD34 accumulation could be observed 0.5 h after intravenous injection of SPIO-antiCD34/NEs. Moreover, the degree of CD133+ EPC accumulation at MI sites was three-fold higher than in control MI model mice, while ensuing microvessel density was roughly two-fold higher than controls and left ventricular ejection fraction was > 50%. Therapeutic cell biodistribution, MI site targeting, and treatment effects were confirmed by SPIO-enhanced MRI. This study offers a new strategy to improve the endogenous EPC-based myocardial ischemic injury repair through NEs mediated SPIO nanoparticle conjugated CD34 antibody delivery and imaging. STATEMENT OF SIGNIFICANCE: The efficacy of endogenous endothelial progenitor cell (EPC)-based cardiovascular repair therapy for ischemic heart damage is limited by relatively low EPC accumulation at the target site. We have developed a method to improve EPC capture by exploiting the strong targeting ability of neutrophils (NEs) to ischemic inflammatory foci and the capacity of these treated cells to release of preloaded cargo with EPC-binding affinity. Briefly, NEs were loaded with superparamagnetic iron oxide nanoparticles conjugated to an antibody against the EPC surface protein CD34 (SPIO-antiCD34). Thus, we explored sites targeting with nanocomposites cargo for non-invasive EPCs interception and therapy tracking. We demonstrate that SPIO-antiCD34 released from NEs can effectively capture endogenous EPCs and thereby promote heart revascularization and functional recovery in mice. Moreover, the entire process can be monitored by SPIO-enhanced magnetic resonance imaging including therapeutic cell biodistribution, myocardial infarction site targeting, and tissue repair.
Collapse
|
24
|
Souza-Neto FV, Islas F, Jiménez-González S, Luaces M, Ramchandani B, Romero-Miranda A, Delgado-Valero B, Roldan-Molina E, Saiz-Pardo M, Cerón-Nieto MÁ, Ortega-Medina L, Martínez-Martínez E, Cachofeiro V. Mitochondrial Oxidative Stress Promotes Cardiac Remodeling in Myocardial Infarction through the Activation of Endoplasmic Reticulum Stress. Antioxidants (Basel) 2022; 11:antiox11071232. [PMID: 35883722 PMCID: PMC9311874 DOI: 10.3390/antiox11071232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
We have evaluated cardiac function and fibrosis in infarcted male Wistar rats treated with MitoQ (50 mg/kg/day) or vehicle for 4 weeks. A cohort of patients admitted with a first episode of acute MI were also analyzed with cardiac magnetic resonance and T1 mapping during admission and at a 12-month follow-up. Infarcted animals presented cardiac hypertrophy and a reduction in the left ventricular ejection fraction (LVEF) and E- and A-waves (E/A) ratio when compared to controls. Myocardial infarction (MI) rats also showed cardiac fibrosis and endoplasmic reticulum (ER) stress activation. Binding immunoglobulin protein (BiP) levels, a marker of ER stress, were correlated with collagen I levels. MitoQ reduced oxidative stress and prevented all these changes without affecting the infarct size. The LVEF and E/A ratio in patients with MI were 57.6 ± 7.9% and 0.96 ± 0.34, respectively. No major changes in cardiac function, extracellular volume fraction (ECV), or LV mass were observed at follow-up. Interestingly, the myeloperoxidase (MPO) levels were associated with the ECV in basal conditions. BiP staining and collagen content were also higher in cardiac samples from autopsies of patients who had suffered an MI than in those who had died from other causes. These results show the interactions between mitochondrial oxidative stress and ER stress, which can result in the development of diffuse fibrosis in the context of MI.
Collapse
Affiliation(s)
- Francisco V. Souza-Neto
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (A.R.-M.); (B.D.-V.)
| | - Fabian Islas
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, 28040 Madrid, Spain; (F.I.); (M.L.)
| | - Sara Jiménez-González
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (A.R.-M.); (B.D.-V.)
| | - María Luaces
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, 28040 Madrid, Spain; (F.I.); (M.L.)
| | - Bunty Ramchandani
- Servicio de Cirugía Cardiaca Infantil, Hospital La Paz, 28046 Madrid, Spain;
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (A.R.-M.); (B.D.-V.)
| | - Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (A.R.-M.); (B.D.-V.)
| | - Elena Roldan-Molina
- Biobanco del Hospital Clínico San Carlos, Instituto de Investigación de Salud del Hospital Clínico San Carlos, 28040 Madrid, Spain; (E.R.-M.); (L.O.-M.)
| | - Melchor Saiz-Pardo
- Departamento de Patología, Hospital Clínico San Carlos, 28040 Madrid, Spain; (M.S.-P.); (M.Á.C.-N.)
- Departamento de Medicina Legal, Psiquiatría y Patología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Mª Ángeles Cerón-Nieto
- Departamento de Patología, Hospital Clínico San Carlos, 28040 Madrid, Spain; (M.S.-P.); (M.Á.C.-N.)
| | - Luis Ortega-Medina
- Biobanco del Hospital Clínico San Carlos, Instituto de Investigación de Salud del Hospital Clínico San Carlos, 28040 Madrid, Spain; (E.R.-M.); (L.O.-M.)
- Departamento de Patología, Hospital Clínico San Carlos, 28040 Madrid, Spain; (M.S.-P.); (M.Á.C.-N.)
- Departamento de Medicina Legal, Psiquiatría y Patología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (A.R.-M.); (B.D.-V.)
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28222 Majadahonda, Spain
- Correspondence: (E.M.-M.); (V.C.); Tel.: +34-91-3941483 (E.M.-M.); +34-91-3941489 (V.C.)
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (A.R.-M.); (B.D.-V.)
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28222 Majadahonda, Spain
- Correspondence: (E.M.-M.); (V.C.); Tel.: +34-91-3941483 (E.M.-M.); +34-91-3941489 (V.C.)
| |
Collapse
|
25
|
O'Brien AT, Gil KE, Varghese J, Simonetti OP, Zareba KM. T2 mapping in myocardial disease: a comprehensive review. J Cardiovasc Magn Reson 2022; 24:33. [PMID: 35659266 PMCID: PMC9167641 DOI: 10.1186/s12968-022-00866-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/27/2022] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular magnetic resonance (CMR) is considered the gold standard imaging modality for myocardial tissue characterization. Elevated transverse relaxation time (T2) is specific for increased myocardial water content, increased free water, and is used as an index of myocardial edema. The strengths of quantitative T2 mapping lie in the accurate characterization of myocardial edema, and the early detection of reversible myocardial disease without the use of contrast agents or ionizing radiation. Quantitative T2 mapping overcomes the limitations of T2-weighted imaging for reliable assessment of diffuse myocardial edema and can be used to diagnose, stage, and monitor myocardial injury. Strong evidence supports the clinical use of T2 mapping in acute myocardial infarction, myocarditis, heart transplant rejection, and dilated cardiomyopathy. Accumulating data support the utility of T2 mapping for the assessment of other cardiomyopathies, rheumatologic conditions with cardiac involvement, and monitoring for cancer therapy-related cardiac injury. Importantly, elevated T2 relaxation time may be the first sign of myocardial injury in many diseases and oftentimes precedes symptoms, changes in ejection fraction, and irreversible myocardial remodeling. This comprehensive review discusses the technical considerations and clinical roles of myocardial T2 mapping with an emphasis on expanding the impact of this unique, noninvasive tissue parameter.
Collapse
Affiliation(s)
- Aaron T O'Brien
- Ohio University Heritage College of Osteopathic Medicine, Athens, Ohio, USA
| | - Katarzyna E Gil
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Juliet Varghese
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Orlando P Simonetti
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Radiology, The Ohio State University, Columbus, Ohio, USA
| | - Karolina M Zareba
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
26
|
Vasques‐Nóvoa F, Angélico‐Gonçalves A, Alvarenga JM, Nobrega J, Cerqueira RJ, Mancio J, Leite‐Moreira AF, Roncon‐Albuquerque R. Myocardial oedema: pathophysiological basis and implications for the failing heart. ESC Heart Fail 2022; 9:958-976. [PMID: 35150087 PMCID: PMC8934951 DOI: 10.1002/ehf2.13775] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/27/2021] [Accepted: 12/02/2021] [Indexed: 12/04/2022] Open
Abstract
Myocardial fluid homeostasis relies on a complex interplay between microvascular filtration, interstitial hydration, cardiomyocyte water uptake and lymphatic removal. Dysregulation of one or more of these mechanisms may result in myocardial oedema. Interstitial and intracellular fluid accumulation disrupts myocardial architecture, intercellular communication, and metabolic pathways, decreasing contractility and increasing myocardial stiffness. The widespread use of cardiac magnetic resonance enabled the identification of myocardial oedema as a clinically relevant imaging finding with prognostic implications in several types of heart failure. Furthermore, growing experimental evidence has contributed to a better understanding of the physical and molecular interactions in the microvascular barrier, myocardial interstitium and lymphatics and how they might be disrupted in heart failure. In this review, we summarize current knowledge on the factors controlling myocardial water balance in the healthy and failing heart and pinpoint the new potential therapeutic avenues.
Collapse
Affiliation(s)
- Francisco Vasques‐Nóvoa
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - António Angélico‐Gonçalves
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - José M.G. Alvarenga
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - João Nobrega
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - Rui J. Cerqueira
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - Jennifer Mancio
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - Adelino F. Leite‐Moreira
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| | - Roberto Roncon‐Albuquerque
- Cardiovascular R&D Center, Faculty of MedicineUniversity of PortoPortoPortugal
- Department of Surgery and Physiology, Faculty of MedicineUniversity of PortoAl. Prof. Hernâni MonteiroPorto4200‐319Portugal
| |
Collapse
|
27
|
Dynamic Trend of Myocardial Edema in Takotsubo Syndrome: A Serial Cardiac Magnetic Resonance Study. J Clin Med 2022; 11:jcm11040987. [PMID: 35207260 PMCID: PMC8878106 DOI: 10.3390/jcm11040987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 11/29/2022] Open
Abstract
Background: The wall motion abnormalities of the left ventricle (LV) in takotsubo syndrome (TTS) are known to be transient and completely recover within a few weeks. However, there is little information about the relationship between functional recovery and tissue characteristics. The aim of this study was to investigate the recovery process of TTS using cardiovascular magnetic resonance (CMR). Methods: Consecutive patients with TTS were prospectively enrolled. We performed serial CMR in the acute phase (<72 h after admission), the subacute phase (7–10 days after admission) and the chronic phase (3 months later). To assess the degree of myocardial edema quantitatively, we evaluated the signal intensity of myocardium on T2-weighted images and calculated the signal intensity ratio compared with the skeletal muscle. Results: Fifteen patients with TTS were enrolled. CMR demonstrated reduced LV ejection fraction in the acute phase, and it recovered almost completely by the subacute phase. On the other hand, severe myocardial edema was still observed in the subacute phase, associated with increased LV mass. The highest signal intensity ratio in the subacute phase was correlated with the maximum voltage of negative T wave on electrocardiogram (r = 0.57, p = 0.03). Conclusions: In patients with TTS, myocardial edema associated with increased LV mass still remained in the subacute phase despite functional recovery of the LV. Electrocardiogram may be useful to assess the degree of myocardial edema in the subacute phase. Our study suggests that myocardial ischemia might have a central role in developing TTS.
Collapse
|
28
|
Ibanez B. Targeting inflammation to improve long-term outcome in ST-segment elevation myocardial infarction survivors. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:124-126. [PMID: 35136996 DOI: 10.1093/ehjacc/zuac002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/11/2022] [Indexed: 06/14/2023]
Affiliation(s)
- Borja Ibanez
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029. Madrid, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| |
Collapse
|
29
|
Berry C, Ibáñez B. Intramyocardial Hemorrhage: The Final Frontier for Preventing Heart Failure Post-Myocardial Infarction. J Am Coll Cardiol 2022; 79:49-51. [PMID: 34991788 DOI: 10.1016/j.jacc.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Colin Berry
- British Heart Foundation Research Centre of Excellence, University of Glasgow, Glasgow, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland.
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares, IIS-Hospital Universitario Fundación Jiménez Díaz, and CIBERCV, Madrid, Spain
| |
Collapse
|
30
|
Moon BF, Iyer SK, Josselyn NJ, Hwuang E, Swago S, Keeney SJ, Castillero E, Ferrari G, Pilla JJ, Gorman JH, Gorman RC, Tschabrunn C, Shou H, Matthai W, Wehrli FW, Ferrari VA, Han Y, Litt H, Witschey WR. Magnetic susceptibility and R2* of myocardial reperfusion injury at 3T and 7T. Magn Reson Med 2022; 87:323-336. [PMID: 34355815 PMCID: PMC9067599 DOI: 10.1002/mrm.28955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Magnetic susceptibility (Δχ) alterations have shown association with myocardial infarction (MI) iron deposition, yet there remains limited understanding of the relationship between relaxation rates and susceptibility or the effect of magnetic field strength. Hence, Δχ and R 2 ∗ in MI were compared at 3T and 7T. METHODS Subacute MI was induced by coronary artery ligation in male Yorkshire swine. 3D multiecho gradient echo imaging was performed at 1-week postinfarction at 3T and 7T. Quantitative susceptibility mapping images were reconstructed using a morphology-enabled dipole inversion. R 2 ∗ maps and quantitative susceptibility mapping were generated to assess the relationship between R 2 ∗ , Δχ, and field strength. Infarct histopathology was investigated. RESULTS Magnetic susceptibility was not significantly different across field strengths (7T: 126.8 ± 41.7 ppb; 3T: 110.2 ± 21.0 ppb, P = NS), unlike R 2 ∗ (7T: 247.0 ± 14.8 Hz; 3T: 106.1 ± 6.5 Hz, P < .001). Additionally, infarct Δχ and R 2 ∗ were significantly higher than remote myocardium. Magnetic susceptibility at 7T versus 3T had a significant association (β = 1.02, R2 = 0.82, P < .001), as did R 2 ∗ (β = 2.35, R2 = 0.98, P < .001). Infarct pathophysiology and iron deposition were detected through histology and compared with imaging findings. CONCLUSION R 2 ∗ showed dependence and Δχ showed independence of field strength. Histology validated the presence of iron and supported imaging findings.
Collapse
Affiliation(s)
- Brianna F. Moon
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Srikant Kamesh Iyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas J. Josselyn
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eileen Hwuang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia Swago
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel J. Keeney
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Estibaliz Castillero
- Department of Surgery, Columbia University Irving Medical Center, New York City, NY, USA
| | - Giovanni Ferrari
- Department of Surgery, Columbia University Irving Medical Center, New York City, NY, USA
| | - James J. Pilla
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph H. Gorman
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert C. Gorman
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cory Tschabrunn
- Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William Matthai
- Department of Medicine, Penn Presbyterian Medical Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix W. Wehrli
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victor A. Ferrari
- Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuchi Han
- Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Harold Litt
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter R. Witschey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Alkhalil M. Novel Applications for Invasive and Non-invasive Tools in the Era of Contemporary Percutaneous Coronary Revascularisation. Curr Cardiol Rev 2022; 18:e190122191004. [PMID: 33530910 PMCID: PMC9241120 DOI: 10.2174/1573403x17666210202102549] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/08/2020] [Accepted: 11/24/2020] [Indexed: 11/22/2022] Open
Abstract
Percutaneous coronary intervention (PCI) is an expanding treatment option for patients with coronary artery disease (CAD). It is considered the default strategy for the unstable presentation of CAD. PCI techniques have evolved over the last 4 decades with significant improvements in stent design, an increase in functional assessment of coronary lesions, and the use of intra-vascular imaging. Nonetheless, the morbidity and mortality related to CAD remain significant. Advances in technology have allowed a better understanding of the nature and progression of CAD. New tools are now available that reflect the pathophysiological changes at the level of the myocardium and coronary atherosclerotic plaque. Certain changes within the plaque would render it more prone to rupture leading to acute vascular events. These changes are potentially detected using novel tools invasively, such as near infra-red spectroscopy, or non-invasively using T2 mapping cardiovascular magnetic resonance imaging (CMR) and 18F-Sodium Fluoride positron emission tomography/ computed tomography. Similarly, changes at the level of the injured myocardium are feasibly assessed invasively using index microcirculatory resistance or non-invasively using T1 mapping CMR. Importantly, these changes could be detected immediately with the opportunity to tailor treatment to those considered at high risk. Concurrently, novel therapeutic options have demonstrated promising results in reducing future cardiovascular risks in patients with CAD. This Review article will discuss the role of these novel tools and their applicability in employing a mechanical and pharmacological treatment to mitigate cardiovascular risk in patients with CAD.
Collapse
Affiliation(s)
- Mohammad Alkhalil
- Department of Cardiothoracic Services, Freeman hospital, Newcastle-upon-Tyne UK.,Department of Cardiology, Toronto General Hospital, Toronto Canada
| |
Collapse
|
32
|
Qian G, A X, Jiang X, Jiang Z, Li T, Dong W, Guo J, Chen Y. Early Trimetazidine Therapy in Patients Undergoing Primary Percutaneous Coronary Intervention for ST Segment Elevation Myocardial Infarction Reduces Myocardial Infarction Size. Cardiovasc Drugs Ther 2021; 37:497-506. [PMID: 34767131 DOI: 10.1007/s10557-021-07259-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Trimetazidine, a metabolic agent with anti-ischemic effects, was reported to reduce reperfusion injury in animal models. In this randomized double-blind placebo-controlled trial, we investigated the effects of trimetazidine on the reduction of infarction size in patients undergoing revascularization for ST segment elevation myocardial infarction (STEMI). METHODS Patients with STEMI randomly received trimetazidine (n = 87) or placebo (n = 86) before primary percutaneous coronary intervention (PCI), and subsequently received oral trimetazidine or placebo for 12 months after reperfusion. The predefined primary endpoint was infarction size on cardiac magnetic resonance (CMR) performed at 7 days after primary PCI. The trial was registered on www.clinicaltrials.gov (registration number: NCT02826616). RESULTS The clinical characteristics of the patients in both groups were well-matched at baseline. At 7 days after primary PCI, the percentage and absolute infarction size in the trimetazidine group were significantly smaller than those in the control group (22% ± 12% [n = 74] vs. 27% ± 13% [n = 74], p = 0.011 and 28 ± 18 g [n = 74] vs. 35 ± 19 g [n = 74], p = 0.022, respectively), and the incidence of myocardial microvascular obstruction (MVO) measured by CMR was significantly reduced in the trimetazidine group (29.7% [22/74] vs. 52.7% [39/74], p = 0.005). The myocardial salvage index (MSI) measured by CMR was significantly higher in the trimetazidine group (48% ± 20% vs. 39% ± 20%, p = 0.008). The incidence of readmission due to aggravated heart failure did not differ significantly between the trimetazidine group and the control group (8.0% vs. 14.0%, p = 0.234). CONCLUSIONS In patients with STEMI undergoing primary PCI, early trimetazidine before reperfusion reduced myocardial infarction size and MVO, and improved MSI.
Collapse
Affiliation(s)
- Geng Qian
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China.
| | - Xin A
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Xiaosi Jiang
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Zichao Jiang
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Tao Li
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Wei Dong
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Jun Guo
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Street, Beijing, 100853, China.
| |
Collapse
|
33
|
Correia C, Wang QD, Linhardt G, Carlsson LG, Ulfenborg B, Walentinsson A, Rydén-Markinhutha K, Behrendt M, Wikström J, Sartipy P, Jennbacken K, Synnergren J. Unraveling the Metabolic Derangements Occurring in Non-infarcted Areas of Pig Hearts With Chronic Heart Failure. Front Cardiovasc Med 2021; 8:753470. [PMID: 34722683 PMCID: PMC8548620 DOI: 10.3389/fcvm.2021.753470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: After myocardial infarction (MI), the non-infarcted left ventricle (LV) ensures appropriate contractile function of the heart. Metabolic disturbance in this region greatly exacerbates post-MI heart failure (HF) pathology. This study aimed to provide a comprehensive understanding of the metabolic derangements occurring in the non-infarcted LV that could trigger cardiovascular deterioration. Methods and Results: We used a pig model that progressed into chronic HF over 3 months following MI induction. Integrated gene and metabolite signatures revealed region-specific perturbations in amino acid- and lipid metabolism, insulin signaling and, oxidative stress response. Remote LV, in particular, showed impaired glutamine and arginine metabolism, altered synthesis of lipids, glucose metabolism disorder, and increased insulin resistance. LPIN1, PPP1R3C, PTPN1, CREM, and NR0B2 were identified as the main effectors in metabolism dysregulation in the remote zone and were found differentially expressed also in the myocardium of patients with ischemic and/or dilated cardiomyopathy. In addition, a simultaneous significant decrease in arginine levels and altered PRCP, PTPN1, and ARF6 expression suggest alterations in vascular function in remote area. Conclusions: This study unravels an array of dysregulated genes and metabolites putatively involved in maladaptive metabolic and vascular remodeling in the non-infarcted myocardium and may contribute to the development of more precise therapies to mitigate progression of chronic HF post-MI.
Collapse
Affiliation(s)
- Cláudia Correia
- Systems Biology Research Center, Translational Bioinformatics Research Group, School of Biosciences, University of Skövde, Skövde, Sweden.,Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gunilla Linhardt
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Leif G Carlsson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Benjamin Ulfenborg
- Systems Biology Research Center, Translational Bioinformatics Research Group, School of Biosciences, University of Skövde, Skövde, Sweden
| | - Anna Walentinsson
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Katarina Rydén-Markinhutha
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Margareta Behrendt
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Johannes Wikström
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter Sartipy
- Systems Biology Research Center, Translational Bioinformatics Research Group, School of Biosciences, University of Skövde, Skövde, Sweden.,Late-Stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jane Synnergren
- Systems Biology Research Center, Translational Bioinformatics Research Group, School of Biosciences, University of Skövde, Skövde, Sweden
| |
Collapse
|
34
|
Francis R, Chong J, Ramlall M, Bucciarelli-Ducci C, Clayton T, Dodd M, Engstrøm T, Evans R, Ferreira VM, Fontana M, Greenwood JP, Kharbanda RK, Kim WY, Kotecha T, Lønborg JT, Mathur A, Møller UK, Moon J, Perkins A, Rakhit RD, Yellon DM, Bøtker HE, Bulluck H, Hausenloy DJ. Effect of remote ischaemic conditioning on infarct size and remodelling in ST-segment elevation myocardial infarction patients: the CONDI-2/ERIC-PPCI CMR substudy. Basic Res Cardiol 2021; 116:59. [PMID: 34648075 PMCID: PMC8516772 DOI: 10.1007/s00395-021-00896-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/06/2021] [Accepted: 09/21/2021] [Indexed: 11/26/2022]
Abstract
The effect of limb remote ischaemic conditioning (RIC) on myocardial infarct (MI) size and left ventricular ejection fraction (LVEF) was investigated in a pre-planned cardiovascular magnetic resonance (CMR) substudy of the CONDI-2/ERIC-PPCI trial. This single-blind multi-centre trial (7 sites in UK and Denmark) included 169 ST-segment elevation myocardial infarction (STEMI) patients who were already randomised to either control (n = 89) or limb RIC (n = 80) (4 × 5 min cycles of arm cuff inflations/deflations) prior to primary percutaneous coronary intervention. CMR was performed acutely and at 6 months. The primary endpoint was MI size on the 6 month CMR scan, expressed as median and interquartile range. In 110 patients with 6-month CMR data, limb RIC did not reduce MI size [RIC: 13.0 (5.1-17.1)% of LV mass; control: 11.1 (7.0-17.8)% of LV mass, P = 0.39], or LVEF, when compared to control. In 162 patients with acute CMR data, limb RIC had no effect on acute MI size, microvascular obstruction and LVEF when compared to control. In a subgroup of anterior STEMI patients, RIC was associated with lower incidence of microvascular obstruction and higher LVEF on the acute scan when compared with control, but this was not associated with an improvement in LVEF at 6 months. In summary, in this pre-planned CMR substudy of the CONDI-2/ERIC-PPCI trial, there was no evidence that limb RIC reduced MI size or improved LVEF at 6 months by CMR, findings which are consistent with the neutral effects of limb RIC on clinical outcomes reported in the main CONDI-2/ERIC-PPCI trial.
Collapse
Affiliation(s)
- Rohin Francis
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Jun Chong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Manish Ramlall
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Chiara Bucciarelli-Ducci
- Biomedical Research Centre, Bristol Heart Institute, National Institute of Health Research (NIHR), University Hospitals Bristol NHS Foundation Trust and University of Bristol, Upper Maudlin St, Bristol, BS2 8HW, UK
| | - Tim Clayton
- London School of Hygiene and Tropical Medicine Clinical Trials Unit, London, UK
| | - Matthew Dodd
- London School of Hygiene and Tropical Medicine Clinical Trials Unit, London, UK
| | - Thomas Engstrøm
- Rigshospitalet, Department of Cardiology, University of Copenhagen, Copenhagen, Denmark
| | - Richard Evans
- London School of Hygiene and Tropical Medicine Clinical Trials Unit, London, UK
| | - Vanessa M Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
- British Heart Foundation Centre of Research Excellence, Oxford, UK
| | - Marianna Fontana
- Royal Free Hospital London and Institute of Cardiovascular Science, University College London, London, UK
| | - John P Greenwood
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Rajesh K Kharbanda
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Won Yong Kim
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of MR Research Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Tushar Kotecha
- Royal Free Hospital London and Institute of Cardiovascular Science, University College London, London, UK
| | - Jacob T Lønborg
- Rigshospitalet, Department of Cardiology, University of Copenhagen, Copenhagen, Denmark
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- William Harvey Research Institute, Queen Mary University London, London, UK
| | - Ulla Kristine Møller
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of MR Research Centre, Aarhus University Hospital, Aarhus, Denmark
| | - James Moon
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Alexander Perkins
- London School of Hygiene and Tropical Medicine Clinical Trials Unit, London, UK
| | - Roby D Rakhit
- Royal Free Hospital London and Institute of Cardiovascular Science, University College London, London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of MR Research Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Heerajnarain Bulluck
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Department of Cardiology, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK.
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.
- Department of Cardiology, National Heart Centre, Singapore, Singapore.
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung, Taiwan.
| |
Collapse
|
35
|
Lorca R, Jiménez-Blanco M, García-Ruiz JM, Pizarro G, Fernández-Jiménez R, García-Álvarez A, Fernández-Friera L, Lobo-González M, Fuster V, Rossello X, Ibáñez B. Coexistencia de progresión transmural y lateral del frente de onda en el infarto de miocardio humano. Rev Esp Cardiol 2021. [DOI: 10.1016/j.recesp.2020.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Stoeck CT, von Deuster C, Fuetterer M, Polacin M, Waschkies CF, van Gorkum RJH, Kron M, Fleischmann T, Cesarovic N, Weisskopf M, Kozerke S. Cardiovascular magnetic resonance imaging of functional and microstructural changes of the heart in a longitudinal pig model of acute to chronic myocardial infarction. J Cardiovasc Magn Reson 2021; 23:103. [PMID: 34538266 PMCID: PMC8451129 DOI: 10.1186/s12968-021-00794-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND We examined the dynamic response of the myocardium to infarction in a longitudinal porcine study using relaxometry, functional as well as diffusion cardiovascular magnetic resonance (CMR). We sought to compare non contrast CMR methods like relaxometry and in-vivo diffusion to contrast enhanced imaging and investigate the link of microstructural and functional changes in the acute and chronically infarcted heart. METHODS CMR was performed on five myocardial infarction pigs and four healthy controls. In the infarction group, measurements were obtained 2 weeks before 90 min occlusion of the left circumflex artery, 6 days after ischemia and at 5 as well as 9 weeks as chronic follow-up. The timing of measurements was replicated in the control cohort. Imaging consisted of functional cine imaging, 3D tagging, T2 mapping, native as well as gadolinium enhanced T1 mapping, cardiac diffusion tensor imaging, and late gadolinium enhancement imaging. RESULTS Native T1, extracellular volume (ECV) and mean diffusivity (MD) were significantly elevated in the infarcted region while fractional anisotropy (FA) was significantly reduced. During the transition from acute to chronic stages, native T1 presented minor changes (< 3%). ECV as well as MD increased from acute to the chronic stages compared to baseline: ECV: 125 ± 24% (day 6) 157 ± 24% (week 5) 146 ± 60% (week 9), MD: 17 ± 7% (day 6) 33 ± 14% (week 5) 29 ± 15% (week 9) and FA was further reduced: - 31 ± 10% (day 6) - 38 ± 8% (week 5) - 36 ± 14% (week 9). T2 as marker for myocardial edema was significantly increased in the ischemic area only during the acute stage (83 ± 3 ms infarction vs. 58 ± 2 ms control p < 0.001 and 61 ± 2 ms in the remote area p < 0.001). The analysis of functional imaging revealed reduced left ventricular ejection fraction, global longitudinal strain and torsion in the infarct group. At the same time the transmural helix angle (HA) gradient was steeper in the chronic follow-up and a correlation between longitudinal strain and transmural HA gradient was detected (r = 0.59 with p < 0.05). Comparing non-gadolinium enhanced data T2 mapping showed the largest relative change between infarct and remote during the acute stage (+ 33 ± 4% day 6, with p = 0.013 T2 vs. MD, p = 0.009 T2 vs. FA and p = 0.01 T2 vs. T1) while FA exhibited the largest relative change between infarct and remote during the chronic follow-up (+ 31 ± 2% week 5, with p = N.S. FA vs. MD, p = 0.03 FA vs. T2 and p = 0.003 FA vs. T1). Overall, diffusion parameters provided a higher contrast (> 23% for MD and > 27% for FA) during follow-up compared to relaxometry (T1 17-18%/T2 10-20%). CONCLUSION During chronic follow-up after myocardial infarction, cardiac diffusion tensor imaging provides a higher sensitivity for mapping microstructural alterations when compared to non-contrast enhanced relaxometry with the added benefit of providing directional tensor information to assess remodelling of myocyte aggregate orientations, which cannot be otherwise assessed.
Collapse
Affiliation(s)
- Christian T. Stoeck
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Constantin von Deuster
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Maximilian Fuetterer
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Malgorzata Polacin
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Conny F. Waschkies
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Robbert J. H. van Gorkum
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Mareike Kron
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Thea Fleischmann
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Nikola Cesarovic
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
- Institute of Translational Cardiovascular Technologies, ETH Zurich, Zurich, Switzerland
| | - Miriam Weisskopf
- Division of Surgical Research, University Hospital Zurich, Zurich, Switzerland
| | - Sebastian Kozerke
- Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| |
Collapse
|
37
|
Saleh Y, Abdelkarim O, Herzallah K, Abela GS. Anthracycline-induced cardiotoxicity: mechanisms of action, incidence, risk factors, prevention, and treatment. Heart Fail Rev 2021; 26:1159-1173. [PMID: 32410142 DOI: 10.1007/s10741-020-09968-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Anthracycline is a mainstay in treatment of many cancers including lymphoma and breast cancer among many others. However, anthracycline treatment can be cardiotoxic. Although anthracycline-induced cardiotoxicity is dose dependent, it can also occur early at the onset of treatment and even up to several years following completion of treatment. This review article focuses on the understanding of mechanisms of anthracycline-induced cardiotoxicity, the treatments, and recommended follow-up and preventive approaches.
Collapse
Affiliation(s)
- Yehia Saleh
- Department of Internal Medicine, Michigan State University, East Lansing, MI, USA
| | - Ola Abdelkarim
- Department of Internal Medicine, Cardiology, Michigan State University, 788 service road, Room B-208, Clinical Center, East Lansing, MI, USA
| | - Khader Herzallah
- Department of Internal Medicine, Michigan State University, East Lansing, MI, USA
| | - George S Abela
- Department of Internal Medicine, Cardiology, Michigan State University, 788 service road, Room B-208, Clinical Center, East Lansing, MI, USA.
| |
Collapse
|
38
|
Beijnink CWH, van der Hoeven NW, Konijnenberg LSF, Kim RJ, Bekkers SCAM, Kloner RA, Everaars H, El Messaoudi S, van Rossum AC, van Royen N, Nijveldt R. Cardiac MRI to Visualize Myocardial Damage after ST-Segment Elevation Myocardial Infarction: A Review of Its Histologic Validation. Radiology 2021; 301:4-18. [PMID: 34427461 DOI: 10.1148/radiol.2021204265] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiac MRI is a noninvasive diagnostic tool using nonionizing radiation that is widely used in patients with ST-segment elevation myocardial infarction (STEMI). Cardiac MRI depicts different prognosticating components of myocardial damage such as edema, intramyocardial hemorrhage (IMH), microvascular obstruction (MVO), and fibrosis. But how do cardiac MRI findings correlate to histologic findings? Shortly after STEMI, T2-weighted imaging and T2* mapping cardiac MRI depict, respectively, edema and IMH. The acute infarct size can be determined with late gadolinium enhancement (LGE) cardiac MRI. T2-weighted MRI should not be used for area-at-risk delineation because T2 values change dynamically over the first few days after STEMI and the severity of T2 abnormalities can be modulated with treatment. Furthermore, LGE cardiac MRI is the most accurate method to visualize MVO, which is characterized by hemorrhage, microvascular injury, and necrosis in histologic samples. In the chronic setting post-STEMI, LGE cardiac MRI is best used to detect replacement fibrosis (ie, final infarct size after injury healing). Finally, native T1 mapping has recently emerged as a contrast material-free method to measure infarct size that, however, remains inferior to LGE cardiac MRI. Especially LGE cardiac MRI-defined infarct size and the presence and extent of MVO may be used to monitor the effect of new therapeutic interventions in the treatment of reperfusion injury and infarct size reduction. © RSNA, 2021 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Casper W H Beijnink
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Nina W van der Hoeven
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Lara S F Konijnenberg
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Raymond J Kim
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Sebastiaan C A M Bekkers
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Robert A Kloner
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Henk Everaars
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Saloua El Messaoudi
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Albert C van Rossum
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Niels van Royen
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| | - Robin Nijveldt
- From the Department of Cardiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands (C.W.H.B., L.S.F.K., S.E.M., N.v.R., R.N.); Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands (N.W.v.d.H., H.E., A.C.v.R.); Department of Medicine, Duke University School of Medicine, Durham, NC (R.J.K.); Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands (S.C.A.M.B.); Huntington Medical Research Institutes, Pasadena, Calif (R.A.K.); and Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, Calif (R.A.K.)
| |
Collapse
|
39
|
Yang MX, Shi K, Xu HY, He Y, Ma M, Zhang L, Wang JL, Li XS, Fu C, Li H, Zhou B, Zhou XY, Yang Z, Guo YK, Yang ZG. Inflammation in Remote Myocardium and Left Ventricular Remodeling After Acute Myocardial Infarction: A Pilot Study Using T2 Mapping. J Magn Reson Imaging 2021; 55:555-564. [PMID: 34245075 DOI: 10.1002/jmri.27827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The pathophysiological changes in the remote myocardium after acute myocardial infarction (MI) remains less understood. PURPOSE To assess the inflammation in the remote myocardium post-MI and its association with left ventricular (LV) remodeling using T2 mapping. STUDY TYPE Prospective. ANIMAL MODEL AND SUBJECTS Twelve pigs at 3-day post-MI, 6 pigs at 3-month post-MI, 6 healthy pigs; 54 patients at 3-day and 3-month post-MI, 31 healthy volunteers; FIELD STRENGTH/SEQUENCE: A 3 T MRI/ steady-state free-precession sequence for T2 mapping (animals: 0, 30, and 55 msec; human: 0, 25, and 55 msec), phase-sensitive inversion recovery gradient echo for late gadolinium enhancement (LGE), balanced steady free-precession sequence for cine. ASSESSMENT Infarcted myocardium was defined on LGE, remote T2 was measured on T2 maps. LV remodeling was evaluated as LV end-diastolic volume change index between two scans using cine. CD68 staining was conducted to detect monocyte/macrophage. STATISTICAL TESTS Student-t test and one-way ANOVA were used to compare remote T2 with normal controls. The association of remote T2 with LV remodeling was assessed using linear regression. P values of <0.05 were used to denote statistical significance. RESULTS Compared with healthy pigs, remote T2 significantly increased from 3 days to 3 months post-MI (31.43 ± 0.67 vs. 33.53 ± 1.15 vs. 36.43 ± 1.07 msec). CD68 staining demonstrated the inflammation in remote myocardium post-MI but not in healthy pigs. Significant remote myocardial alterations in T2 were also observed in human group (40.51 ± 1.79 vs. 41.94 ± 1.14 vs. 42.52 ± 1.71 msec). In patients, the 3-month remote T2 (β = 0.432) and remote T2 variation between two scans (β = 0.554) were both independently associated with LV remodeling. CONCLUSION T2 mapping could characterize the abnormalities in the remote myocardium post-MI, which was potentially caused by the inflammatory response. Moreover, variations in remote T2 were associated with LV remodeling. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Meng-Xi Yang
- Department of Radiology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Ke Shi
- Department of Radiology, West China Hospital, Sichuan University, Sichuan, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Yong He
- Department of Cardiology, West China Hospital, Sichuan University, Sichuan, China
| | - Min Ma
- Department of Cardiology, West China Hospital, Sichuan University, Sichuan, China.,Department of Cardiology, The Sixth People's Hospital of Chengdu, Sichuan, China
| | - Lu Zhang
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | | | - Xue-Sheng Li
- Department of Radiology, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Chuan Fu
- Department of Radiology, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Hong Li
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Bin Zhou
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Xiao-Yue Zhou
- MR Collaboration, Siemens Healthcare Ltd, Shanghai, China
| | - Zhi Yang
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China.,Department of Radiology, Chengdu Fifth People's Hospital, Sichuan, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
40
|
Masci PG, Pavon AG, Pontone G, Symons R, Lorenzoni V, Francone M, Zalewski J, Barison A, Guglielmo M, Aquaro GD, Galea N, Muscogiuri G, Muller O, Carbone I, Baggiano A, Iglesias JF, Nessler J, Andreini D, Camici PG, Claus P, de Luca L, Agati L, Janssens S, Schwitter J, Bogaert J. Early or deferred cardiovascular magnetic resonance after ST-segment-elevation myocardial infarction for effective risk stratification. Eur Heart J Cardiovasc Imaging 2021; 21:632-639. [PMID: 31326993 DOI: 10.1093/ehjci/jez179] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/04/2019] [Accepted: 06/20/2019] [Indexed: 11/14/2022] Open
Abstract
AIMS In ST-segment-elevation myocardial infarction (STEMI), cardiovascular magnetic resonance (CMR) holds the potentiality to improve risk stratification in addition to Thrombolysis in Myocardial Infarction (TIMI) risk score. Nevertheless, the optimal timing for CMR after STEMI remains poorly defined. We aim at comparing the prognostic performance of three stratification strategies according to the timing of CMR after STEMI. METHODS AND RESULTS The population of this prospective registry-based study included 492 reperfused STEMI patients. All patients underwent post-reperfusion (median: 4 days post-STEMI) and follow-up (median: 4.8 months post-STEMI) CMR. Left ventricular (LV) volumes, function, infarct size, and microvascular obstruction extent were quantified. Primary endpoint was a composite of all-death and heart failure (HF) hospitalization. Baseline-to-follow-up percentage increase of LV end-diastolic (EDV; ΔLV-EDV) ≥20% or end-systolic volumes (ESV; ΔLV-ESV) ≥15% were tested against outcome. Three multivariate models were developed including TIMI risk score plus early post-STEMI (early-CMR) or follow-up CMR (deferred-CMR) or both CMRs parameters along with adverse LV remodelling (paired-CMRs). During a median follow-up of 8.3 years, the primary endpoint occurred in 84 patients (47 deaths; 37 HF hospitalizations). Early-CMR, deferred-CMR, and paired-CMR demonstrated similar predictive value for the primary endpoint (C-statistic: 0.726, 0.728, and 0.738, respectively; P = 0.663). ΔLV-EDV ≥20% or ΔLV-ESV ≥15% were unadjusted outcome predictors (hazard ratio: 2.020 and 2.032, respectively; P = 0.002 for both) but lost their predictive value when corrected for other covariates in paired-CMR model. CONCLUSION In STEMI patients, early-, deferred-, or paired-CMR were equivalent stratification strategies for outcome prediction. Adverse LV remodelling parameters were not independent prognosticators.
Collapse
Affiliation(s)
- Pier Giorgio Masci
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital Campus, Westminster Bridge Road, London SE1 7EH, UK
| | - Anna Giulia Pavon
- Cardiology Division, Heart & Vessels Department, Center of Cardiac Magnetic Resonance, Rue du Bugnon 46, 1005 Lausanne, University Hospital, Lausanne, Switzerland
| | - Gianluca Pontone
- Centro Cardiologico Monzino, IRCCS Via Carlo Parea, 4, 20138 Milan, Italy
| | - Rolf Symons
- Radiology Department, Gasthuisberg University Hospitals, Herestraat 49, 3000 Leuven, Belgium
| | - Valentina Lorenzoni
- Institute of Management, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 24, 56127 Pisa, Italy
| | - Marco Francone
- Department of Radiological, Oncological, and Pathological Sciences, La Sapienza University, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Jaroslaw Zalewski
- Department of Coronary Disease, Jagiellonian University Medical College, Gołębia 24, 31-007 Kraków, Poland.,Department of Interventional Cardiology, John Paul II Hospital, Prądnicka 80, 31-202 Kraköw, Poland
| | - Andrea Barison
- Fondazione CNR-Regione Toscana 'G.Monasterio', Via Moruzzi 1, 56100 Pisa, Italy
| | - Marco Guglielmo
- Centro Cardiologico Monzino, IRCCS Via Carlo Parea, 4, 20138 Milan, Italy
| | | | - Nicola Galea
- Department of Radiological, Oncological, and Pathological Sciences, La Sapienza University, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | | | - Olivier Muller
- Cardiology Division, Heart & Vessels Department, Center of Cardiac Magnetic Resonance, Rue du Bugnon 46, 1005 Lausanne, University Hospital, Lausanne, Switzerland
| | - Iacopo Carbone
- Department of Radiological, Oncological, and Pathological Sciences, La Sapienza University, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Andrea Baggiano
- Radiology Department, Gasthuisberg University Hospitals, Herestraat 49, 3000 Leuven, Belgium
| | - Juan F Iglesias
- Cardiology Division, University Hospitals Geneve, Rue Gabrielle-Perret-Gentil 4, 1205 Genève, Switzerland
| | - Jadwiga Nessler
- Department of Coronary Disease, Jagiellonian University Medical College, Gołębia 24, 31-007 Kraków, Poland
| | - Daniele Andreini
- Centro Cardiologico Monzino, IRCCS Via Carlo Parea, 4, 20138 Milan, Italy
| | - Paolo G Camici
- Cardiology Division, Heart & Vessels Department, Center of Cardiac Magnetic Resonance, Rue du Bugnon 46, 1005 Lausanne, University Hospital, Lausanne, Switzerland
| | - Piet Claus
- Cardiology Department, Gasthuisberg University Hospitals, Herestraat 49, 3000 Leuven, Belgium
| | - Laura de Luca
- Cardiology Department, La Sapienza University, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Luciano Agati
- Cardiology Department, La Sapienza University, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Stefan Janssens
- Cardiology Department, Gasthuisberg University Hospitals, Herestraat 49, 3000 Leuven, Belgium
| | - Jurg Schwitter
- Cardiology Division, Heart & Vessels Department, Center of Cardiac Magnetic Resonance, Rue du Bugnon 46, 1005 Lausanne, University Hospital, Lausanne, Switzerland
| | - Jan Bogaert
- Radiology Department, Gasthuisberg University Hospitals, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
41
|
Guo Z, Zhao M, Jia G, Ma R, Li M. LncRNA PART1 alleviated myocardial ischemia/reperfusion injury via suppressing miR-503-5p/BIRC5 mediated mitochondrial apoptosis. Int J Cardiol 2021; 338:176-184. [PMID: 34082009 DOI: 10.1016/j.ijcard.2021.05.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Long non-coding RNA (lncRNA) is crucial for heart development and for adult heart structural maintenance and function. Herein, we performed a study to explore the effect of lncRNA PART1 on myocardial ischemia-reperfusion (I/R) injury by targeting BIRC5 through miR-503-5p pathway. METHODS I/R model was created in vivo and vitro. The level of gene and protein was detected by RT-PCR and western blot. The apoptosis level was assessed by TUNEL and flow cytometry. Cell viability was determined by MTT. Mitochondrial function was evaluated by ATP content, ROS production, GSH level, and mitochondrial membrane potential. Cardiac function was confirmed by echocardiography, TTC staining, and H&E staining. RESULTS Here, we found that the expression of lncRNA PART1 was down-regulated in the I/R hearts and H/R cardiomyocytes. Forced expression of PART1 remitted cardiac I/RI and H/R cardiomyocyte injury. Silencing of PART1 aggravated apoptosis and mitochondrial damage in cardiomyocytes. We found that PART1 functioned as a competing endogenous RNA of miR-503-5p, which decreased the expression of miR-503-5p. We further established BIRC5 as a target of miR-503-5p. Furthermore, PART1 prevented apoptosis and improved mitochondrial function in myocardial I/RI by targeting miR-503-5p/BIRC5. CONCLUSIONS In summary, PART1 protected mitochondrial function via miR-503-5p/BIRC5 pathway in MI/RI, which may provide the new theoretical basis for MI/RI treatment in the clinic.
Collapse
Affiliation(s)
- Zhihao Guo
- Department of Cardiology III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Ming Zhao
- Department of Cardiology III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China.
| | - Guowei Jia
- Department of Cardiology III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Rui Ma
- Department of Cardiology III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Meili Li
- Department of Cardiology III, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| |
Collapse
|
42
|
Citric Acid Cycle Metabolites Predict Infarct Size in Pigs Submitted to Transient Coronary Artery Occlusion and Treated with Succinate Dehydrogenase Inhibitors or Remote Ischemic Perconditioning. Int J Mol Sci 2021; 22:ijms22084151. [PMID: 33923786 PMCID: PMC8072915 DOI: 10.3390/ijms22084151] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/02/2022] Open
Abstract
Succinate dehydrogenase (SDH) inhibition with malonate during reperfusion reduced myocardial infarction in animals, whereas its endogenous substrate, succinate, is detected in plasma from STEMI patients. We investigated whether protection by SDH inhibition is additive to that of remote ischemic perconditioning (RIC) in pigs submitted to transient coronary artery occlusion, and whether protective maneuvers influence plasma levels of citric acid cycle metabolites. Forty pigs were submitted to 40 min coronary occlusion and reperfusion, and allocated to four groups (controls, sodium malonate 10 mmol/L, RIC, and malonate + RIC). Plasma was obtained from femoral and great cardiac veins and analyzed by LC-MS/MS. Malonate, RIC, and malonate + RIC reduced infarct size (24.67 ± 5.98, 25.29 ± 3.92 and 29.83 ± 4.62% vs. 46.47 ± 4.49% in controls, p < 0.05), but no additive effects were detected. Enhanced concentrations of succinate, fumarate, malate and citrate were observed in controls during initial reperfusion in the great cardiac vein, and most were reduced by cardioprotective maneuvers. Concentrations of succinate, fumarate, and malate significantly correlated with infarct size. In conclusion, despite the combination of SDH inhibition during reperfusion and RIC did not result in additive protection, plasma concentrations of selected citric acid cycle metabolites are attenuated by protective maneuvers, correlate with irreversible injury, and might become a prognosis tool in STEMI patients.
Collapse
|
43
|
Silvis MJM, van Hout GPJ, Fiolet ATL, Dekker M, Bosch L, van Nieuwburg MMJ, Visser J, Jansen MS, Timmers L, de Kleijn DPV. Experimental parameters and infarct size in closed chest pig LAD ischemia reperfusion models; lessons learned. BMC Cardiovasc Disord 2021; 21:171. [PMID: 33845779 PMCID: PMC8042863 DOI: 10.1186/s12872-021-01995-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/05/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Preclinical models that resemble the clinical setting as closely as possible are essential in translating promising therapies for the treatment of acute myocardial infarction. Closed chest pig left anterior descending coronary artery (LAD) ischemia reperfusion (I/R) models are valuable and clinically relevant. Knowledge on the influence of experimental design on infarct size (IS) in these models is a prerequisite for suitable models. To this end, we investigated the impact of several experimental features (occlusion and follow-up time and influence of area at risk (AAR)) on IS. METHODS A total of fifty-one female Landrace pigs were subjected to closed chest LAD balloon occlusion and evaluated in three substudies with varying protocols. To assess the relationship between time of occlusion and the IS, 18 pigs were subjected to 60-, 75- and 90 min of occlusion and terminated after 24 h of follow-up. Influence of prolonged follow-up on IS was studied in 18 pigs after 75 min of occlusion that were terminated at 1, 3 and 7 days. The relation between AAR and IS was studied in 28 pigs after 60 min of occlusion and 24 h of follow-up. The relation between VF, number of shocks and IS was studied in the same 28 pigs after 60 min of occlusion. RESULTS Increasing occlusion time resulted in an increased IS as a ratio of the AAR (IS/AAR). This ranged from 53 ± 23% after 60 min of occlusion to 88 ± 2.2% after 90 min (P = 0.01). Increasing follow-up, from 1 to 3 or 7 days after 75 min of occlusion did not effect IS/AAR. Increasing AAR led to a larger IS/AAR (r2 = 0.34, P = 0.002), earlier VF (r2 = 0.32, P = 0.027) and a higher number of shocks (r2 = 0.29, P = 0.004) in pigs subjected to 60 min of occlusion. CONCLUSIONS These experiments describe the association of occlusion time, follow-up duration, AAR and VF with IS in closed chest pig LAD I/R models. These results have important implications for future I/R studies in pigs and can serve as a guideline for the selection of appropriate parameters and the optimal experimental design.
Collapse
Affiliation(s)
- Max J M Silvis
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerardus P J van Hout
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Aernoud T L Fiolet
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mirthe Dekker
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Vascular Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Lena Bosch
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Joyce Visser
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marlijn S Jansen
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leo Timmers
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Dominique P V de Kleijn
- Department of Vascular Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands.
| |
Collapse
|
44
|
Detection of Myocardial Infarction by Cardiac Magnetic Resonance in Embolic Stroke Related to First Diagnosed Atrial Fibrillation. J Stroke Cerebrovasc Dis 2021; 30:105753. [PMID: 33845423 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Elevated troponin levels are found in a significant number of patients who are diagnosed with acute embolic stroke (AES) after first diagnosed atrial fibrillation (AF). These myocardial injuries, which are known as cardiocerebral infarction (CCI), are potentially caused by coronary embolism and correspond to simultaneous cardiac and cerebral embolisms. However, this severe condition remains poorly understood. In this prospective study, we aimed to investigate the prevalence and the cardiac magnetic resonance (CMR) characteristics of CCI. MATERIALS AND METHODS Consecutive patients with first diagnosed AF hospitalized for AES in a neurovascular intensive care unit from 2019 to 2020 were included. Troponin Ic kinetic were measured <72 h, MRI and coronary angiography or CT scan were performed <7 days after admission. Patients with significant coronary lesions were excluded. RESULTS During the study period, 1150 patients with strokes were hospitalized in the neurovascular intensive care unit (ICU). Of these patients, 955 had an ischemic stroke and 97 had a transient ischemic attack. Among the 44 patients with AES and with first diagnosed AF, 34 patients underwent CMR and CMR analysis identified 12 MI. A significant rise in troponin (>0.10 µg/L) was observed in 35% of the total population (12/34 patients). More specifically, a rise was seen in 23% of the AES without MI group, 58% of the AES with MI. In addition, coronary embolism was identified in 3 patients who underwent coronary angiography (3/12) and MI was often (30%) localized in infero-latero-medial and infero-apical segments. Most AES were localized in the superficial sylvian territory. CONCLUSION We found a high prevalence of CMR-confirmed double embolization sites in the acute phase of an embolic stroke. Further studies are required to better characterize the pathophysiology, clinical course and prognostic value of CCI. Moreover, optimal management strategies, including antiplatelet therapy, remain to be determined.
Collapse
|
45
|
Gómez-Talavera S, Fernandez-Jimenez R, Fuster V, Nothnagel ND, Kouwenhoven M, Clemence M, García-Lunar I, Gómez-Rubín MC, Navarro F, Pérez-Asenjo B, Fernández-Friera L, Calero MJ, Orejas M, Cabrera JA, Desco M, Pizarro G, Ibáñez B, Sánchez-González J. Clinical Validation of a 3-Dimensional Ultrafast Cardiac Magnetic Resonance Protocol Including Single Breath-Hold 3-Dimensional Sequences. JACC Cardiovasc Imaging 2021; 14:1742-1754. [PMID: 33865783 PMCID: PMC8421247 DOI: 10.1016/j.jcmg.2021.02.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/05/2021] [Accepted: 02/05/2021] [Indexed: 11/02/2022]
Abstract
OBJECTIVES This study sought to clinically validate a novel 3-dimensional (3D) ultrafast cardiac magnetic resonance (CMR) protocol including cine (anatomy and function) and late gadolinium enhancement (LGE), each in a single breath-hold. BACKGROUND CMR is the reference tool for cardiac imaging but is time-consuming. METHODS A protocol comprising isotropic 3D cine (Enhanced sensitivity encoding [SENSE] by Static Outer volume Subtraction [ESSOS]) and isotropic 3D LGE sequences was compared with a standard cine+LGE protocol in a prospective study of 107 patients (age 58 ± 11 years; 24% female). Left ventricular (LV) mass, volumes, and LV and right ventricular (RV) ejection fraction (LVEF, RVEF) were assessed by 3D ESSOS and 2D cine CMR. LGE (% LV) was assessed using 3D and 2D sequences. RESULTS Three-dimensional and LGE acquisitions lasted 24 and 22 s, respectively. Three-dimensional and LGE images were of good quality and allowed quantification in all cases. Mean LVEF by 3D and 2D CMR were 51 ± 12% and 52 ± 12%, respectively, with excellent intermethod agreement (intraclass correlation coefficient [ICC]: 0.96; 95% confidence interval [CI]: 0.94 to 0.97) and insignificant bias. Mean RVEF 3D and 2D CMR were 60.4 ± 5.4% and 59.7 ± 5.2%, respectively, with acceptable intermethod agreement (ICC: 0.73; 95% CI: 0.63 to 0.81) and insignificant bias. Both 2D and 3D LGE showed excellent agreement, and intraobserver and interobserver agreement were excellent for 3D LGE. CONCLUSIONS ESSOS single breath-hold 3D CMR allows accurate assessment of heart anatomy and function. Combining ESSOS with 3D LGE allows complete cardiac examination in <1 min of acquisition time. This protocol expands the indication for CMR, reduces costs, and increases patient comfort.
Collapse
Affiliation(s)
- Sandra Gómez-Talavera
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Department of Cardiology, IIS-Hospital Fundacion Jiménez Díaz, Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Rodrigo Fernandez-Jimenez
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Department of Cardiology, Hospital Universitario Clinico San Carlos, Madrid, Spain
| | - Valentín Fuster
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | - Inés García-Lunar
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Department of Cardiology, Hospital Universitario Quiron UEM, Madrid, Spain
| | | | - Felipe Navarro
- Department of Cardiology, IIS-Hospital Fundacion Jiménez Díaz, Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Braulio Pérez-Asenjo
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Leticia Fernández-Friera
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Department of Cardiology, Hospital Montepríncipe-CEU, Madrid, Spain
| | - María J Calero
- Department of Cardiology, Hospital Universtario Rey Juan Carlos-Móstoles, Madrid, Spain
| | - Miguel Orejas
- Department of Cardiology, IIS-Hospital Fundacion Jiménez Díaz, Madrid, Spain
| | - José A Cabrera
- Department of Cardiology, Hospital Universitario Quiron UEM, Madrid, Spain
| | - Manuel Desco
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aerospacial, Universidad Carlos III, Madrid, Spain
| | - Gonzalo Pizarro
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Department of Cardiology, Hospital Universitario Quiron UEM, Madrid, Spain
| | - Borja Ibáñez
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Department of Cardiology, IIS-Hospital Fundacion Jiménez Díaz, Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | | |
Collapse
|
46
|
Variations in T2-Mapping-Assessed Area at Risk After Experimental Ischemia/Reperfusion. J Cardiovasc Transl Res 2021; 14:1040-1042. [PMID: 33733431 DOI: 10.1007/s12265-021-10120-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/02/2021] [Indexed: 10/21/2022]
|
47
|
Intestinal ischemic reperfusion injury: Recommended rats model and comprehensive review for protective strategies. Biomed Pharmacother 2021; 138:111482. [PMID: 33740527 DOI: 10.1016/j.biopha.2021.111482] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/01/2021] [Accepted: 03/06/2021] [Indexed: 12/17/2022] Open
Abstract
Intestinal ischemic reperfusion injury (IIRI) is a life-threatening condition with high morbidity and mortality in the clinic. IIRI was induced by intestinal ischemic diseases such as, small bowel transplantation, aortic aneurysm surgery, and strangulated hernias. Although related mechanisms have not been fully elucidated, during the last decade, researches have demonstrated that many factors are crucial in the pathological process, including oxidative stress (OS), epithelial barrier function disorder, and so on. Rats model, as the most applied animal IIRI model, provides specific targets for researches and therapeutic strategies. Moreover, various treatment strategies such as, anti-oxidative stress, anti-apoptosis, and anti-inflammation, have shown promising effects in alleviating IIRI. However, current researches cannot solve the clinical problems of IIRI, and specific treatment strategies are still needed to be exploited. This review focuses on a recommended experimental IIRI rat model and understanding of the involved mechanisms such as, OS, gut bacteria translocation, apoptosis, and necroptosis, aim at providing novel ideas for therapeutic strategies of IIRI.
Collapse
|
48
|
Bøtker HE. Searching myocardial rescue through intermittent upper arm occlusion and lizard saliva. Basic Res Cardiol 2021; 116:5. [PMID: 33495904 DOI: 10.1007/s00395-021-00843-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/26/2023]
Affiliation(s)
- Hans Erik Bøtker
- Faculty of Health, Aarhus University, Vennelyst Boulevard 4, 8000, Aarhus C, Denmark.
| |
Collapse
|
49
|
Effect of COMBinAtion therapy with remote ischemic conditioning and exenatide on the Myocardial Infarct size: a two-by-two factorial randomized trial (COMBAT-MI). Basic Res Cardiol 2021; 116:4. [PMID: 33495853 DOI: 10.1007/s00395-021-00842-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/04/2021] [Indexed: 01/03/2023]
Abstract
Remote ischemic conditioning (RIC) and the GLP-1 analog exenatide activate different cardioprotective pathways and may have additive effects on infarct size (IS). Here, we aimed to assess the efficacy of RIC as compared with sham procedure, and of exenatide, as compared with placebo, and the interaction between both, to reduce IS in humans. We designed a two-by-two factorial, randomized controlled, blinded, multicenter, clinical trial. Patients with ST-segment elevation myocardial infarction receiving primary percutaneous coronary intervention (PPCI) within 6 h of symptoms were randomized to RIC or sham procedure and exenatide or matching placebo. The primary outcome was IS measured by late gadolinium enhancement in cardiac magnetic resonance performed 3-7 days after PPCI. The secondary outcomes were myocardial salvage index, transmurality index, left ventricular ejection fraction and relative microvascular obstruction volume. A total of 378 patients were randomly allocated, and after applying exclusion criteria, 222 patients were available for analysis. There were no significant interactions between the two randomization factors on the primary or secondary outcomes. IS was similar between groups for the RIC (24 ± 11.8% in the RIC group vs 23.7 ± 10.9% in the sham group, P = 0.827) and the exenatide hypotheses (25.1 ± 11.5% in the exenatide group vs 22.5 ± 10.9% in the placebo group, P = 0.092). There were no effects with either RIC or exenatide on the secondary outcomes. Unexpected adverse events or side effects of RIC and exenatide were not observed. In conclusion, neither RIC nor exenatide, or its combination, were able to reduce IS in STEMI patients when administered as an adjunct to PPCI.
Collapse
|
50
|
Shi K, Yang MX, Xia CC, Peng WL, Zhang K, Li ZL, Guo YK, Yang ZG. Noninvasive oxygenation assessment after acute myocardial infarction with breathing maneuvers-induced oxygenation-sensitive magnetic resonance imaging. J Magn Reson Imaging 2021; 54:284-289. [PMID: 33433045 DOI: 10.1002/jmri.27509] [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: 11/23/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 02/05/2023] Open
Abstract
The safety profiles when performing stress oxygenation-sensitive magnetic resonance imaging (OS-MRI) have raised concerns in clinical practice. Adenosine infusion can cause side effects such as chest pain, dyspnea, arrhythmia, and even cardiac death. The aim of this study was to investigate the feasibility of breathing maneuvers-induced OS-MRI in acute myocardial infarction (MI). This was a prospective study, which included 14 healthy rabbits and nine MI rabbit models. This study used 3 T MRI/modified Look-Locker inversion recovery sequence for native T1 mapping, balanced steady-state free precession sequence for OS imaging, and phase-sensitive inversion recovery sequence for late gadolinium enhancement. The changes in myocardial oxygenation (ΔSI) were assessed under two breathing maneuvers protocols in healthy rabbits: a series of extended breath-holding (BH), and a combined maneuver of hyperventilation followed by the extended BH (HVBH). Subsequently, OS-MRI with HVBH in acute MI rabbits was performed, and the ΔSI was compared with that of adenosine stress protocol. Student's t-test, Wilcoxon rank test, and Friedman test were used to compare ΔSI in different subgroups. Pearson and Spearman correlation was used to obtain the association of ΔSI between breathing maneuvers and adenosine stress. Bland-Altman analysis was used to assess the bias of ΔSI between HVBH and adenosine stress. In healthy rabbits, BH maneuvers from 30 to 50 s induced significant increase in SI compared with the baseline (all p < 0.05). By contrast, hyperventilation for 60 s followed by 10 s-BH (HVBH 10 s) exhibited a comparable ΔSI to that of stress test (p = 0.07). In acute MI rabbits, HVBH 10 s-induced ΔSIs among infarcted, salvaged, and the remote myocardial area were no less effectiveness than adenosine stress when performing OS-MRI (r = 0.84; p < 0.05). Combined breathing maneuvers with OS-MRI have the potential to be used as a nonpharmacological alternative for assessing myocardial oxygenation in patients with acute MI. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Ke Shi
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meng-Xi Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Department of Radiology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chun-Chao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wan-Lin Peng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kun Zhang
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhen-Lin Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhi-Gang Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|