151
|
Comparison of 0.9-mm and 1.4-mm catheters in excimer laser coronary angioplasty for acute myocardial infarction. Lasers Med Sci 2019; 34:1747-1754. [PMID: 30879227 DOI: 10.1007/s10103-019-02772-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Excimer laser coronary angioplasty (ELCA) is a unique revascularization modality that can vaporize plaque and thrombus. Compared to thrombus aspiration therapy, ELCA is reported to provide better microcirculation and reduced peripheral embolism in treatment for acute coronary syndrome. Excimer laser catheters come in various sizes, and we sought to compare the 0.9- and 1.4-mm-diameter catheters. We retrospectively enrolled 90 acute myocardial infarction (AMI) patients who underwent primary percutaneous coronary intervention with excimer laser from August 2013 to March 2017 in our hospital. Patients were grouped by diameter of catheter that had been used, the 0.9 mm group (n = 51) and 1.4 mm group (n = 39). We evaluated myocardial perfusion, procedural success rate, short-term outcome, lesion crossability, and complications between the two groups. The percentage of patients whose final thrombolysis in myocardial infarction (TIMI) grade was 3 (0.9 mm 86.3% vs 1.4 mm 89.7% p = 0.75) and final myocardial blush grade (MBG) was 3 (0.9 mm 72.5% vs 1.4 mm 69.2% p = 0.82) was similarly high for both groups. Procedural success rate, in-hospital major adverse cardiac events (MACE), lesion crossability, and complications were also similar. This study showed that efficacy of 0.9 and 1.4 mm excimer laser catheter was equivalent in ELCA for AMI patients. If one takes into account lesion crossability, debulking effects, and the stunned platelets phenomenon, the 0.9 mm excimer laser catheter is sufficient for ELCA in AMI patients.
Collapse
|
152
|
Borlotti A, Jerosch-Herold M, Liu D, Viliani D, Bracco A, Alkhalil M, De Maria GL, Channon KM, Banning AP, Choudhury RP, Neubauer S, Kharbanda RK, Dall'Armellina E. Acute Microvascular Impairment Post-Reperfused STEMI Is Reversible and Has Additional Clinical Predictive Value: A CMR OxAMI Study. JACC Cardiovasc Imaging 2019; 12:1783-1793. [PMID: 30660541 PMCID: PMC6718360 DOI: 10.1016/j.jcmg.2018.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/21/2022]
Abstract
Objectives This study sought to investigate the clinical utility and the predictive relevance of absolute rest myocardial blood flow (MBF) by cardiac magnetic resonance (CMR) in acute myocardial infarction. Background Microvascular obstruction (MVO) remains one of the worst prognostic factors in patients with reperfused ST-segment elevation myocardial infarction (STEMI). Clinical trials have focused on cardioprotective strategies to maintain microvascular functionality, but there is a need for a noninvasive test to determine their efficacy. Methods A total of 64 STEMI patients post–primary percutaneous coronary intervention underwent 3-T CMR scans acutely and at 6 months (6M). The protocol included cine function, T2-weighted edema imaging, pre-contrast T1 mapping, rest first-pass perfusion, and late gadolinium enhancement imaging. Segmental MBF, corrected for rate pressure product (MBFcor), was quantified in remote, edematous, and infarcted myocardium. Results Acute MBFcor was significantly reduced in infarcted myocardium compared with remote MBF (MBFinfarct 0.76 ± 0.20 ml/min/g vs. MBFremote 1.02 ± 0.21 ml/min/g, p < 0.001), but it significantly increased at 6M (MBFinfarct 0.76 ± 0.20 ml/min/g acute vs. 0.85 ± 0.22 ml/min/g at 6M, p < 0.001). On a segmental basis, acute MBFcor had incremental prognostic value for infarct size at 6M (odds of no LGE at 6M increased by 1.4:1 [p < 0.001] for each 0.1 ml/min/g increase of acute MBFcor) and functional recovery (odds of wall thickening >45% at 6M increased by 1.38:1 [p < 0.001] for each 0.1 ml/min/g increase of acute MBFcor). In subjects with coronary flow reserve >2 or index of myocardial resistance <40, acute MBF was associated with long-term functional recovery and was an independent predictor of infarct size reduction. Conclusions Acute MBF by CMR could represent a novel quantitative imaging biomarker of microvascular reversibility, and it could be used to identify patients who may benefit from more intensive or novel therapies.
Collapse
Affiliation(s)
- Alessandra Borlotti
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Michael Jerosch-Herold
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dan Liu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Dafne Viliani
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alessia Bracco
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mohammad Alkhalil
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Giovanni Luigi De Maria
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | - Keith M Channon
- Oxford Heart Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Adrian P Banning
- Oxford Heart Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | - Erica Dall'Armellina
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
153
|
The presence of late potentials after percutaneous coronary intervention for the treatment of acute coronary syndrome as a predictor for future significant cardiac events resulting in re-hospitalization. J Electrocardiol 2019; 53:71-78. [PMID: 30703576 DOI: 10.1016/j.jelectrocard.2019.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/18/2018] [Accepted: 01/01/2019] [Indexed: 01/03/2023]
Abstract
INTRODUCTION We previously reported that LP positive patients after percutaneous coronary intervention (PCI) had higher rate of re-hospitalization in the small-scale study (135 patients). In this study, we evaluated correlation between LP and later cardiac events leading to re-hospitalization more extensively in greater population. METHODS AND RESULTS A 24-h high-resolution (HR) ambulatory electrocardiogram (ECG) was performed in 421 patients that received PCI for the treatment of acute coronary syndrome (ACS) within 30 days. Various baseline characteristics and post-PCI ECG parameters including LP were examined for correlation with later re-hospitalization. LP was evaluated based on 3 different conditions, i.e., the worst, mean and best values, from 24-h signal-averaged QRS wave data. During the post-PCI follow-up period (611 ± 489.0 days), 90 patients were re-hospitalized due to cardiac events. Multivariate analysis identified only positive LP based on the worst value as an independent predictor for re-hospitalization with OR 2.26. Most of re-hospitalization cases (>75%) were predominantly attributed to ischemic events. LP positive population had significantly higher incidences of ischemic events as well as overall re-hospitalization compared to LP negative population. The predictive power of LP was decreased when it was combined with other variables. The receiver operating characteristic analysis determined the LP cut-off values consistent with the LP positive criteria previously reported and standardized. CONCLUSION The presence of LP in the 24-h HR ambulatory ECG post-PCI was an independent predictor for a risk of re-hospitalization due to ischemic cardiac events in ACS patients.
Collapse
|
154
|
McCartney PJ, Eteiba H, Maznyczka AM, McEntegart M, Greenwood JP, Muir DF, Chowdhary S, Gershlick AH, Appleby C, Cotton JM, Wragg A, Curzen N, Oldroyd KG, Lindsay M, Rocchiccioli JP, Shaukat A, Good R, Watkins S, Robertson K, Malkin C, Martin L, Gillespie L, Ford TJ, Petrie MC, Macfarlane PW, Tait RC, Welsh P, Sattar N, Weir RA, Fox KA, Ford I, McConnachie A, Berry C. Effect of Low-Dose Intracoronary Alteplase During Primary Percutaneous Coronary Intervention on Microvascular Obstruction in Patients With Acute Myocardial Infarction: A Randomized Clinical Trial. JAMA 2019; 321:56-68. [PMID: 30620371 PMCID: PMC6583564 DOI: 10.1001/jama.2018.19802] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
IMPORTANCE Microvascular obstruction commonly affects patients with acute ST-segment elevation myocardial infarction (STEMI) and is associated with adverse outcomes. OBJECTIVE To determine whether a therapeutic strategy involving low-dose intracoronary fibrinolytic therapy with alteplase infused early after coronary reperfusion will reduce microvascular obstruction. DESIGN, SETTING, AND PARTICIPANTS Between March 17, 2016, and December 21, 2017, 440 patients presenting at 11 hospitals in the United Kingdom within 6 hours of STEMI due to a proximal-mid-vessel occlusion of a major coronary artery were randomized in a 1:1:1 dose-ranging trial design. Patient follow-up to 3 months was completed on April 12, 2018. INTERVENTIONS Participants were randomly assigned to treatment with placebo (n = 151), alteplase 10 mg (n = 144), or alteplase 20 mg (n = 145) by manual infusion over 5 to 10 minutes. The intervention was scheduled to occur early during the primary PCI procedure, after reperfusion of the infarct-related coronary artery and before stent implant. MAIN OUTCOMES AND MEASURES The primary outcome was the amount of microvascular obstruction (% left ventricular mass) demonstrated by contrast-enhanced cardiac magnetic resonance imaging (MRI) conducted from days 2 through 7 after enrollment. The primary comparison was the alteplase 20-mg group vs the placebo group; if not significant, the alteplase 10-mg group vs the placebo group was considered a secondary analysis. RESULTS Recruitment stopped on December 21, 2017, because conditional power for the primary outcome based on a prespecified analysis of the first 267 randomized participants was less than 30% in both treatment groups (futility criterion). Among the 440 patients randomized (mean age, 60.5 years; 15% women), the primary end point was achieved in 396 patients (90%), 17 (3.9%) withdrew, and all others were followed up to 3 months. In the primary analysis, the mean microvascular obstruction did not differ between the 20-mg alteplase and placebo groups (3.5% vs 2.3%; estimated difference, 1.16%; 95% CI, -0.08% to 2.41%; P = .32) nor in the analysis of 10-mg alteplase vs placebo groups (2.6% vs 2.3%; estimated difference, 0.29%; 95% CI, -0.76% to 1.35%; P = .74). Major adverse cardiac events (cardiac death, nonfatal MI, unplanned hospitalization for heart failure) occurred in 15 patients (10.1%) in the placebo group, 18 (12.9%) in the 10-mg alteplase group, and 12 (8.2%) in the 20-mg alteplase group. CONCLUSIONS AND RELEVANCE Among patients with acute STEMI presenting within 6 hours of symptoms, adjunctive low-dose intracoronary alteplase given during the primary percutaneous intervention did not reduce microvascular obstruction. The study findings do not support this treatment. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02257294.
Collapse
Affiliation(s)
- Peter J. McCartney
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| | - Hany Eteiba
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| | - Annette M. Maznyczka
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| | - Margaret McEntegart
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| | - John P. Greenwood
- Leeds University and Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Douglas F. Muir
- James Cook University Hospital NHS Trust, Middlesbrough, United Kingdom
| | - Saqib Chowdhary
- South Manchester Hospitals NHS Trust, Manchester, United Kingdom
| | | | - Clare Appleby
- Liverpool Heart and Chest Hospital NHS Foundation Trust, Liverpool, United Kingdom
| | - James M. Cotton
- Royal Wolverhampton University Hospital NHS Trust, Wolverhampton, United Kingdom
| | - Andrew Wragg
- Barts and the London Hospital, London, United Kingdom
| | - Nick Curzen
- University Hospital Southampton Foundation Trust, Southampton, United Kingdom
| | - Keith G. Oldroyd
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Mitchell Lindsay
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - J. Paul Rocchiccioli
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Aadil Shaukat
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Richard Good
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Stuart Watkins
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Keith Robertson
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Christopher Malkin
- Leeds University and Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Lynn Martin
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| | | | - Thomas J. Ford
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Mark C. Petrie
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| | - Peter W. Macfarlane
- Electrocardiography Core Laboratory, University of Glasgow, Glasgow, United Kingdom
| | - R. Campbell Tait
- Department of Haematology, Royal Infirmary, Glasgow, United Kingdom
| | - Paul Welsh
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Naveed Sattar
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Robin A. Weir
- University Hospital Hairmyres, East Kilbride, United Kingdom
| | - Keith A. Fox
- University of Edinburgh, Edinburgh, United Kingdom
| | - Ian Ford
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Alex McConnachie
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Colin Berry
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Clydebank, United Kingdom
| |
Collapse
|
155
|
Microvascular obstruction in non-infarct related coronary arteries is an independent predictor of major adverse cardiovascular events in patients with ST segment-elevation myocardial infarction. Int J Cardiol 2018; 273:22-28. [DOI: 10.1016/j.ijcard.2018.08.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/27/2018] [Accepted: 08/08/2018] [Indexed: 11/15/2022]
|
156
|
Mangion K, Carrick D, Carberry J, Mahrous A, McComb C, Oldroyd KG, Eteiba H, Lindsay M, McEntegart M, Hood S, Petrie MC, Watkins S, Davie A, Zhong X, Epstein FH, Haig CE, Berry C. Circumferential Strain Predicts Major Adverse Cardiovascular Events Following an Acute ST-Segment-Elevation Myocardial Infarction. Radiology 2018; 290:329-337. [PMID: 30457480 DOI: 10.1148/radiol.2018181253] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Purpose To investigate the prognostic value of circumferential left ventricular (LV) strain measured by using cardiac MRI for prediction of major adverse cardiac events (MACE) following an acute ST-segment-elevation myocardial infarction (STEMI). Materials and Methods Participants with acute STEMI were prospectively enrolled from May 11, 2011, to November 22, 2012. Cardiac MRI was performed at 1.5 T during the index hospitalization. Displacement encoding with stimulated echoes (DENSE) and feature tracking of cine cardiac MRI was used to assess circumferential LV strain. MACE that occurred after discharge were independently assessed by cardiologists blinded to the baseline observations. Results A total of 259 participants (mean age, 58 years ± 11 [standard deviation]; 198 men [mean age, 58 years ± 11] and 61 women [mean age, 58 years ± 12]) underwent cardiac MRI 2.2 days ± 1.9 after STEMI. Average infarct size was 18% ± 13 of LV mass and circumferential strain was -13% ± 3 (DENSE method) and -24% ± 7 (feature- tracking method). Fifty-one percent (131 of 259 participants) had presence of microvascular obstruction. During a median follow-up period of 4 years, 8% (21 of 259) experienced MACE. Area under the curve (AUC) for DENSE was different from that of feature tracking (AUC, 0.76 vs 0.62; P = .03). AUC for DENSE was similar to that of initial infarct size (P = .06) and extent of microvascular obstruction (P = .08). DENSE-derived strain provided incremental prognostic benefit over infarct size for prediction of MACE (hazard ratio, 1.3; P < .01). Conclusion Circumferential strain has independent prognostic importance in study participants with acute ST-segment-elevation myocardial infarction. Published under a CC BY 4.0 license. Online supplemental material is available for this article. See also the editorial by Kramer in this issue.
Collapse
Affiliation(s)
- Kenneth Mangion
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - David Carrick
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Jaclyn Carberry
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Ahmed Mahrous
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Christie McComb
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Keith G Oldroyd
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Hany Eteiba
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Mitchell Lindsay
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Margaret McEntegart
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Stuart Hood
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Mark C Petrie
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Stuart Watkins
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Andrew Davie
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Xiaodong Zhong
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Frederick H Epstein
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Caroline E Haig
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| | - Colin Berry
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences (K.M., D.C., J.C., C.M., M.C.P., C.B.), and Robertson Centre for Biostatistics (C.E.H.), University of Glasgow, 126 University Place, Glasgow G12 8TA, Scotland; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Scotland (K.M., D.C., A.M., K.G.O., H.E., M.L., M.M., S.H., M.C.P., S.W., A.D., C.B.); Department of Clinical Physics, NHS Greater Glasgow and Clyde, Glasgow, Scotland (C.M.); Department of MR R&D Collaborations, Siemens Healthcare, Atlanta, Ga (X.Z.); and Department of Biomedical Engineering, University of Virginia, Charlottesville, Va (F.H.E.)
| |
Collapse
|
157
|
Matsuura R, Miyagawa S, Fukushima S, Goto T, Harada A, Shimozaki Y, Yamaki K, Sanami S, Kikuta J, Ishii M, Sawa Y. Intravital imaging with two-photon microscopy reveals cellular dynamics in the ischeamia-reperfused rat heart. Sci Rep 2018; 8:15991. [PMID: 30375442 PMCID: PMC6207786 DOI: 10.1038/s41598-018-34295-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 10/09/2018] [Indexed: 12/27/2022] Open
Abstract
Recent advances in intravital microscopy have provided insight into dynamic biological events at the cellular level in both healthy and pathological tissue. However, real-time in vivo cellular imaging of the beating heart has not been fully established, mainly due to the difficulty of obtaining clear images through cycles of cardiac and respiratory motion. Here we report the successful recording of clear in vivo moving images of the beating rat heart by two-photon microscopy facilitated by cardiothoracic surgery and a novel cardiac stabiliser. Subcellular dynamics of the major cardiac components including the myocardium and its subcellular structures (i.e., nuclei and myofibrils) and mitochondrial distribution in cardiac myocytes were visualised for 4-5 h in green fluorescent protein-expressing transgenic Lewis rats at 15 frames/s. We also observed ischaemia/reperfusion (I/R) injury-induced suppression of the contraction/relaxation cycle and the consequent increase in cell permeability and leukocyte accumulation in cardiac tissue. I/R injury was induced in other transgenic mouse lines to further clarify the biological events in cardiac tissue. This imaging system can serve as an alternative modality for real time monitoring in animal models and cardiological drug screening, and can contribute to the development of more effective treatments for cardiac diseases.
Collapse
Affiliation(s)
- Ryohei Matsuura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satsuki Fukushima
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takasumi Goto
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuri Shimozaki
- Research and Development Division for Advanced Technology, Research and Development Center, Dai Nippon Printing Co., Ltd., Tokyo, Japan
| | - Kazumasa Yamaki
- Research and Development Division for Advanced Technology, Research and Development Center, Dai Nippon Printing Co., Ltd., Tokyo, Japan
| | - Sho Sanami
- Research and Development Division for Advanced Technology, Research and Development Center, Dai Nippon Printing Co., Ltd., Tokyo, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
158
|
González-Montero J, Brito R, Gajardo AIJ, Rodrigo R. Myocardial reperfusion injury and oxidative stress: Therapeutic opportunities. World J Cardiol 2018; 10:74-86. [PMID: 30344955 PMCID: PMC6189069 DOI: 10.4330/wjc.v10.i9.74] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/27/2018] [Accepted: 05/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of death worldwide. Its associated mortality, morbidity and complications have significantly decreased with the development of interventional cardiology and percutaneous coronary angioplasty (PCA) treatment, which quickly and effectively restore the blood flow to the area previously subjected to ischemia. Paradoxically, the restoration of blood flow to the ischemic zone leads to a massive production of reactive oxygen species (ROS) which generate rapid and severe damage to biomolecules, generating a phenomenon called myocardial reperfusion injury (MRI). In the clinical setting, MRI is associated with multiple complications such as lethal reperfusion, no-reflow, myocardial stunning, and reperfusion arrhythmias. Despite significant advances in the understanding of the mechanisms accounting for the myocardial ischemia reperfusion injury, it remains an unsolved problem. Although promising results have been obtained in experimental studies (mainly in animal models), these benefits have not been translated into clinical settings. Thus, clinical trials have failed to find benefits from any therapy to prevent MRI. There is major evidence with respect to the contribution of oxidative stress to MRI in cardiovascular diseases. The lack of consistency between basic studies and clinical trials is not solely based on the diversity inherent in epidemiology but is also a result of the methodological weaknesses of some studies. It is quite possible that pharmacological issues, such as doses, active ingredients, bioavailability, routes of administration, co-therapies, startup time of the drug intervention, and its continuity may also have some responsibility for the lack of consistency between different studies. Furthermore, the administration of high ascorbate doses prior to reperfusion appears to be a safe and rational therapy against the development of oxidative damage associated with myocardial reperfusion. In addition, the association with N-acetylcysteine (a glutathione donor) and deferoxamine (an iron chelator) could improve the antioxidant cardioprotection by ascorbate, making it even more effective in preventing myocardial reperfusion damage associated with PCA following AMI.
Collapse
Affiliation(s)
- Jaime González-Montero
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
| | - Roberto Brito
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
- Internal Medicine Department, University of Chile, Clinical Hospital, Santiago 70058, Chile
| | - Abraham IJ Gajardo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
- Internal Medicine Department, University of Chile, Clinical Hospital, Santiago 70058, Chile
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
| |
Collapse
|
159
|
Chen T, Vunjak-Novakovic G. In vitro Models of Ischemia-Reperfusion Injury. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:142-153. [PMID: 30393757 PMCID: PMC6208331 DOI: 10.1007/s40883-018-0056-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/25/2018] [Indexed: 01/23/2023]
Abstract
Timely reperfusion after a myocardial infarction is necessary to salvage the ischemic region; however, reperfusion itself is also a major contributor to the final tissue damage. Currently, there is no clinically relevant therapy available to reduce ischemia-reperfusion injury (IRI). While many drugs have shown promise in reducing IRI in preclinical studies, none of these drugs have demonstrated benefit in large clinical trials. Part of this failure to translate therapies can be attributed to the reliance on small animal models for preclinical studies. While animal models encapsulate the complexity of the systemic in vivo environment, they do not fully recapitulate human cardiac physiology. Furthermore, it is difficult to uncouple the various interacting pathways in vivo. In contrast, in vitro models using isolated cardiomyocytes allow studies of the direct effect of therapeutics on cardiomyocytes. External factors can be controlled in simulated ischemia-reperfusion to allow for better understanding of the mechanisms that drive IRI. In addition, the availability of cardiomyocytes derived from human induced pluripotent stem cells (hIPS-CMs) offers the opportunity to recapitulate human physiology in vitro. Unfortunately, hIPS-CMs are relatively fetal in phenotype, and are more resistant to hypoxia than the mature cells. Tissue engineering platforms can promote cardiomyocyte maturation for a more predictive physiologic response. These platforms can further be improved upon to account for the heterogenous patient populations seen in the clinical settings and facilitate the translation of therapies. Thereby, the current preclinical studies can be further developed using currently available tools to achieve better predictive drug testing and understanding of IRI. In this article, we discuss the state of the art of in vitro modeling of IRI, propose the roles for tissue engineering in studying IRI and testing the new therapeutic modalities, and how the human tissue models can facilitate translation into the clinic.
Collapse
Affiliation(s)
- Timothy Chen
- Department of Biomedical Engineering, University in the City of New York
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, University in the City of New York
- Department of Medicine Columbia University in the City of New York
| |
Collapse
|
160
|
|
161
|
Abstract
The no-reflow phenomenon refers to the observation that when an organ is made ischemic by occlusion of a large artery supplying it, restoration of patency in that artery does not restore perfusion to the microvasculature supplying the parenchyma of that organ. This has been observed after prolonged arterial occlusions in the heart (30–90 min), brain, skin, and kidney. In experimental models, zones of no reflow in the heart are characterized by ultrastructural microvascular damage, including focal endothelial swelling obstructing the lumen of small vessels. Blood elements such as neutrophil plugs, platelets, and stacking of erythrocytes have also been implicated. No reflow is associated with poor healing of the myocardial infarction. In patients, no reflow is associated with a poor clinical outcome independent of infarct size, suggesting that therapy for no reflow may be an important approach to improving outcome for ST elevation myocardial infarction. No reflow occurs after reperfusion of experimental cerebral ischemia and may be observed after only 5-min episodes of ischemia. Aggregation of blood elements may play a greater role than in cardiac no reflow. No reflow in the brain may involve cortical spreading depression with disturbed local vascular control and high, vasculotonic levels of extracellular K+ concentration, postischemic swelling in endothelial cells and abutting end feet of pericytes, pericyte contraction and death, interstitial edema with collapse of cerebral capillaries, and inflammatory reaction. New guidelines suggesting that reperfusion for stroke may be considered as late as 24 h after the onset of symptoms suggest that clinicians may be seeing more no reflow in the future.
Collapse
Affiliation(s)
- Robert A. Kloner
- Huntington Medical Research Institutes, Pasadena, California
- Cardiovascular Division, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kevin S. King
- Huntington Medical Research Institutes, Pasadena, California
| | - Michael G. Harrington
- Huntington Medical Research Institutes, Pasadena, California
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
162
|
Reinstadler SJ, Stiermaier T, Reindl M, Feistritzer HJ, Fuernau G, Eitel C, Desch S, Klug G, Thiele H, Metzler B, Eitel I. Intramyocardial haemorrhage and prognosis after ST-elevation myocardial infarction. Eur Heart J Cardiovasc Imaging 2018; 20:138-146. [DOI: 10.1093/ehjci/jey101] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/19/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Sebastian J Reinstadler
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstraße 35, Innsbruck, Austria
| | - Thomas Stiermaier
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| | - Martin Reindl
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstraße 35, Innsbruck, Austria
| | - Hans-Josef Feistritzer
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstraße 35, Innsbruck, Austria
| | - Georg Fuernau
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| | - Charlotte Eitel
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| | - Steffen Desch
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| | - Gert Klug
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstraße 35, Innsbruck, Austria
| | - Holger Thiele
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| | - Bernhard Metzler
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstraße 35, Innsbruck, Austria
| | - Ingo Eitel
- Department of Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, Medical Clinic II, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, Lübeck, Germany
| |
Collapse
|
163
|
Chen BH, Shi RY, An DA, Wu R, Wu CW, Hu J, Manly A, Kaddurah H, He J, Pu J, Xu JR, Wu LM. BOLD cardiac MRI for differentiating reversible and irreversible myocardial damage in ST segment elevation myocardial infarction. Eur Radiol 2018; 29:951-962. [PMID: 29987418 DOI: 10.1007/s00330-018-5612-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/07/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES BOLD imaging is a quantitative MRI technique allowing the evaluation of the balance between supply/demand in myocardial oxygenation and myocardial haemorrhage. We sought to investigate the ability of BOLD imaging to differentiate reversible from irreversible myocardial injury as well as the chronological progression of myocardial oxygenation after reperfusion in patients with ST segment elevation myocardial infarction (STEMI). METHODS Twenty-two patients (age, 60 ± 11 years; 77.3% male) with STEMI underwent cardiac MRIs on four occasions: on days 1, 3, 7 and 30 after reperfusion. BOLD MRI was obtained with a multi-echo turbo field echo (TFE) sequence on a 3-T scanner to assess myocardial oxygenation in MI. RESULTS T2* value in MI with intramyocardial haemorrhage (IMH) was the lowest (9.77 ± 3.29 ms), while that of the salvaged zone was the highest (33.97 ± 3.42 ms). T2* values in salvaged myocardium demonstrated a unimodal temporal pattern from days 1 (37.91 ± 2.23 ms) to 30 (30.68 ± 1.59 ms). T2* values in the MI regions were significantly lower than those in remote myocardium, although the trends in both were constant overall. There was a slightly positive correlation between T2* in MI regions and EF (Rho = 0.27, p < 0.05) or SV (Rho = 0.22, p = 0.04) and a slightly negative correlation between T2* in salvaged myocardium and LVEDV (Rho = - 0.23, p < 0.05). CONCLUSIONS BOLD MRI performed in post-STEMI patients allows accurate evaluation of myocardial damage severity and could differentiate reversible from irreversible myocardial injury. The increased T2* values may imply the pathophysiological mechanism of salvaged myocardium. BOLD MRI could represent a more accurate alternative to the other currently available options. KEY POINTS • Myocardial oxygenation and haemorrhage after myocardial infarction affect BOLD MRI values • BOLD MRI could be used to differentiate irreversible from reversible myocardial damage • Changed oxygenation implies the pathophysiological mechanism of salvaged myocardium.
Collapse
Affiliation(s)
- Bing-Hua Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Ruo-Yang Shi
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Dong-Aolei An
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Rui Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Chong-Wen Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Amanda Manly
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Hisham Kaddurah
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Jie He
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jian-Rong Xu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China.
| | - Lian-Ming Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
164
|
Abstract
PURPOSE OF REVIEW Coronary artery no-reflow phenomenon is an incidental outcome of percutaneous coronary intervention in patients presenting with acute myocardial infarction. Despite advances in pharmacologic and non-pharmacologic therapies, coronary no-reflow phenomenon occurs more commonly than desired. It often results in poor clinical outcomes and remains as a relevant consideration in the cardiac catheterization laboratory. In this systematic review, we have sought to discuss the topic in detail, and to relay the most recent discoveries and data on management of this condition. RECENT FINDINGS We discuss several pharmacologic and non-pharmacologic treatments used in the prevention and management of coronary no-reflow and microvascular obstruction. Covered topics include the understanding of pharmacologic mechanisms of current and future agents, and recent discoveries that may result in the development of future treatment options. We conclude that the pathophysiology of coronary no-reflow phenomenon and microvascular obstruction still remains incompletely understood, although several plausible theories have led to the current standard of care for its management. We also conclude that coronary no-reflow phenomenon and microvascular obstruction must be recognized as a multifactorial condition that has certain predispositions and characteristics, therefore its prevention and treatment must begin pre-procedurally and be multi-faceted including certain medications and operator techniques in the cardiac catheterization laboratory.
Collapse
Affiliation(s)
- Ahmadreza Karimianpour
- Department of Cardiovascular Diseases, Heart & Vascular Institute, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA.
| | - Anbukarasi Maran
- Department of Cardiovascular Diseases, Heart & Vascular Institute, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA
| |
Collapse
|
165
|
Reperfusing the myocardium - a damocles Sword. Indian Heart J 2018; 70:433-438. [PMID: 29961464 PMCID: PMC6034085 DOI: 10.1016/j.ihj.2017.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/03/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
Abstract
Return of blood flow after periodic ischemia is often accompanied by myocardial injury, commonly known as lethal reperfusion injury (RI). Experimental studies have shown that 50% of muscle die of ischemia and another 50% die because of reperfusion. It is characterized by myocardial, vascular, or electrophysiological dysfunction that is induced by the restoration of blood flow to previously ischemic tissue. This phenomenon reduces the efficiency of the present modalities used to combat the ischemic myocardium. Moreover, despite an improved understanding of the pathophysiology of this process and encouraging preclinical trials of multiple agents, most of the clinical trials to prevent RI have been disappointing and leaves us at ground zero to explore newer approaches.
Collapse
|
166
|
Schram HCF, Hemradj VV, Hermanides RS, Kedhi E, Ottervanger JP. Coronary artery ectasia, an independent predictor of no-reflow after primary PCI for ST-elevation myocardial infarction. Int J Cardiol 2018; 265:12-17. [PMID: 29731349 DOI: 10.1016/j.ijcard.2018.04.120] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND The no-reflow phenomenon is a serious complication after primary percutaneous coronary intervention (PCI) for ST-elevation Myocardial Infarction (STEMI). Coronary artery ectasia (CAE) may increase the risk of no-reflow, however, only limited data is available on the potential impact of CAE. The aim of this study was to determine the potential association between CAE and no-reflow after primary PCI. METHODS A case control study was performed based on a prospective cohort of STEMI patients from January 2000 to December 2011. All patients with TIMI 0-1 flow post primary PCI, in the absence of dissection, thrombus, spasm or high-grade residual stenosis, were considered as no-reflow case. Control subjects were two consecutive STEMI patients after each case, with TIMI flow ≥2 after primary PCI. CAE was defined as dilatation of an arterial segment to a diameter at least 1.5 times that of the adjacent normal coronary artery. RESULTS In the no-reflow group, frequency of CAE was significantly higher (33.8% vs 3.9%, p < 0.001) compared to the control group. Baseline variables were comparable between patients with and without CAE. Patients with CAE had more often TIMI 0-1 flow pre-PCI (91% vs 71% p = 0.03), less often anterior STEMI (3% vs 37%, p < 0.001) and underwent significantly less often a PCI with stenting (47% vs 74%, p = 0.003). After multivariate analysis, CAE remained a strong and independent predictor of no-reflow (OR 13.9, CI 4.7-41.2, p < 0.001). CONCLUSION CAE is a strong and independent predictor of no-reflow after primary PCI for STEMI. Future studies should assess optimal treatment.
Collapse
Affiliation(s)
| | | | | | - E Kedhi
- Isala, Zwolle, The Netherlands
| | | | -
- Isala, Zwolle, The Netherlands
| |
Collapse
|
167
|
Kloner RA, Dai W, Hale SL. No-Reflow Phenomenon. A New Target for Therapy of Acute Myocardial Infarction Independent of Myocardial Infarct Size. J Cardiovasc Pharmacol Ther 2018; 23:273-276. [DOI: 10.1177/1074248418764467] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Robert A. Kloner
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
- Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA, USA
| | - Wangde Dai
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
- Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA, USA
| | - Sharon L. Hale
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
| |
Collapse
|
168
|
Contrast agent dose and slow/no-reflow in percutaneous coronary interventions : A case-control study of patients with non-ST-segment elevation acute coronary syndromes. Herz 2018; 44:69-75. [PMID: 29637230 DOI: 10.1007/s00059-018-4692-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND The angiographic slow/no-reflow phenomenon after primary percutaneous coronary intervention carries a poor prognosis for patients with non-ST-segment elevation acute coronary syndrome (NSTEACS). There is evidence that contrast agents cause endothelial dysfunction, myocardial cell damage, and coronary spasms. We hypothesized that the contrast agent dose may be related to slow/no-reflow in patients with NSTEACS undergoing percutaneous transluminal coronary angioplasty and stent (PTCA + stent). PATIENTS AND METHODS We enrolled 3369 patients with NSTEACS who underwent PTCA + stent only in the culprit vessel for the first time from September 2007 to May 2017 in this study. Coronary blood flow of ≤TIMI grade 2 after PTCA + stent was defined as slow/no-reflow. The relationship between the contrast agent dose and slow/no-reflow phenomenon was analyzed by multivariate conditional logistic regression and smooth curve fitting. RESULTS In multivariable conditional logistic regression analysis, the contrast agent dose was found to be an independent risk factor for slow/no-reflow after adjusting for the number of stents and the thrombus burden (OR: 1.0112; 95% CI: 1.0049-1.0176; p < 0.0001), and after adjusting for type 2 diabetes mellitus, NSTEACS risk stratification, application of platelet glycoprotein (GP) IIB/IIIA receptor antagonists, type of contrast agent, number of balloon dilatations, number of stents, and thrombus burden (OR: 1.0113; 95% CI: 1.0036-1.0191; p = 0.004). Further, the risk of slow/no-reflow increased significantly with the contrast agent level up to the inflection point of 160 ml. CONCLUSION The contrast agent dose may be a risk factor for slow/no-reflow phenomenon after PTCA + stent in patients with NSTEACS. When the dose was greater than 160 ml, the risk of slow/no-reflow increased significantly.
Collapse
|
169
|
Makepeace CM, Suarez-Pierre A, Kanter EM, Schuessler RB, Nichols CG, Lawton JS. Superior diastolic function with K ATP channel opener diazoxide in a novel mouse Langendorff model. J Surg Res 2018; 227:186-193. [PMID: 29804852 DOI: 10.1016/j.jss.2018.02.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/11/2018] [Accepted: 02/14/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND Adenosine triphosphate-sensitive potassium (KATP) channel openers have been found to be cardioprotective in multiple animal models via an unknown mechanism. Mouse models allow genetic manipulation of KATP channel components for the investigation of this mechanism. Mouse Langendorff models using 30 min of global ischemia are known to induce measurable myocardial infarction and injury. Prolongation of global ischemia in a mouse Langendorff model could allow the determination of the mechanisms involved in KATP channel opener cardioprotection. METHODS Mouse hearts (C57BL/6) underwent baseline perfusion with Krebs-Henseleit buffer (30 min), assessment of function using a left ventricular balloon, delivery of test solution, and prolonged global ischemia (90 min). Hearts underwent reperfusion (30 min) and functional assessment. Coronary flow was measured using an inline probe. Test solutions included were as follows: hyperkalemic cardioplegia alone (CPG, n = 11) or with diazoxide (CPG + DZX, n = 12). RESULTS Although the CPG + DZX group had greater percent recovery of developed pressure and coronary flow, this was not statistically significant. Following a mean of 74 min (CPG) and 77 min (CPG + DZX), an additional increase in end-diastolic pressure was noted (plateau), which was significantly higher in the CPG group. Similarly, the end-diastolic pressure (at reperfusion and at the end of experiment) was significantly higher in the CPG group. CONCLUSIONS Prolongation of global ischemia demonstrated added benefit when DZX was added to traditional hyperkalemic CPG. This model will allow the investigation of DZX mechanism of cardioprotection following manipulation of targeted KATP channel components. This model will also allow translation to prolonged ischemic episodes associated with cardiac surgery.
Collapse
Affiliation(s)
- Carol M Makepeace
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Alejandro Suarez-Pierre
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Evelyn M Kanter
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Richard B Schuessler
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri; Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer S Lawton
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
170
|
Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, Li Y, Zhou H, Chen Y. Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol 2018; 16:157-168. [PMID: 29502045 PMCID: PMC5952878 DOI: 10.1016/j.redox.2018.02.019] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 12/16/2022] Open
Abstract
Receptor-interacting protein 3 (Ripk3)-mediated necroptosis contributes to cardiac ischaemia-reperfusion (IR) injury through poorly defined mechanisms. Our results demonstrated that Ripk3 was strongly upregulated in murine hearts subjected to IR injury and cardiomyocytes treated with LPS and H2O2. The higher level of Ripk3 was positively correlated to the infarction area expansion, cardiac dysfunction and augmented cardiomyocytes necroptosis. Function study further illustrated that upregulated Ripk3 evoked the endoplasmic reticulum (ER) stress, which was accompanied with an increase in intracellular Ca2+ level ([Ca2+]c) and xanthine oxidase (XO) expression. Activated XO raised cellular reactive oxygen species (ROS) that mediated the mitochondrial permeability transition pore (mPTP) opening and cardiomyocytes necroptosis. By comparison, genetic ablation of Ripk3 abrogated the ER stress and thus blocked the [Ca2+]c overload-XO-ROS-mPTP pathways, favouring a pro-survival state that ultimately resulted in the inhibition of cardiomyocytes necroptosis in the setting of cardiac IR injury. In summary, the present study helps to elucidate how necroptosis is mediated by ER stress, via the calcium overload /XO/ROS/mPTP opening axis. ER stress is activated by Ripk3 in cardiac IR injury. ER stress induces calcium overload which triggers XO-dependent ROS overproduction. ROS outburst promotes mPTP opening that accounts for the necroptosis. Inhibiting ER stress favors cardiomyocytes survival and protects cardiac function.
Collapse
Affiliation(s)
- Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qinhua Jin
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Dandan Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Sam Toan
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA 92521 USA
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071 USA.
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
171
|
Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J 2018; 38:774-784. [PMID: 27354052 DOI: 10.1093/eurheartj/ehw224] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022] Open
Abstract
The incidence of ST segment elevation myocardial infarction (STEMI) has decreased over the last two decades in developed countries, but mortality from STEMI despite widespread access to reperfusion therapy is still substantial as is the development of heart failure, particularly among an expanding older population. In developing countries, the incidence of STEMI is increasing and interventional reperfusion is often not available. We here review the pathophysiology of acute myocardial infarction and reperfusion, notably the temporal and spatial evolution of ischaemic and reperfusion injury, the different modes of cell death, and the resulting coronary microvascular dysfunction. We then go on to briefly characterize the cardioprotective phenomena of ischaemic preconditioning, ischaemic postconditioning, and remote ischaemic conditioning and their underlying signal transduction pathways. We discuss in detail the attempts to translate conditioning strategies and drug therapy into the clinical setting. Most attempts have failed so far to reduce infarct size and improve clinical outcomes in STEMI patients, and we discuss potential reasons for such failure. Currently, it appears that remote ischaemic conditioning and a few drugs (atrial natriuretic peptide, exenatide, metoprolol, and esmolol) reduce infarct size, but studies with clinical outcome as primary endpoint are still underway.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, 45122 Essen, Germany
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
172
|
Yu SJ, Buerck JP, O'Rear EA, Whitsett TL. Possible erythrocyte contributions to and exacerbation of the post-thrombolytic no-reflow phenomenon. Biorheology 2018; 54:81-93. [PMID: 29332028 DOI: 10.3233/bir-17144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Reperfusion injury often occurs with therapeutic intervention addressing the arterial occlusions causing acute myocardial infarction and stroke. The no-reflow phenomenon has been ascribed to leukocyte plugging and blood vessel constriction in the microcirculation. OBJECTIVE To assess possible red cell contributions to post-thrombolytic no-reflow phenomenon. METHODS Blood clots were formed by recalcifying 1 ml of citrated fresh human venous blood and then lysed by adding 1,000 units of streptokinase (SK) at several intervals within 1 hour. Red cell deformability was tested by both a microscopic photometric and a filtration technique, viscosity by a cone and plate viscometer, and erythrocyte aggregation by an optical aggregometer. RESULTS Two sampling methods were devised for the microscopic photometric test, both of which indicated increases of erythrocyte stiffness after being lysed from the clot by SK. In accompanying experiments, the viscosity, aggregation and filterability of the post-lytic erythrocytes were assessed. Results indicated increased viscosity in Ringer's, decreased aggregation index and filterability through a 5 μm pore size Nuclepore membrane. CONCLUSION Findings demonstrated that post-lytic changes in red cell deformability do occur which could contribute to the no-reflow phenomenon.
Collapse
Affiliation(s)
- Shang-Jen Yu
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - James P Buerck
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Edgar A O'Rear
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Thomas L Whitsett
- Department of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK 73014, USA
| |
Collapse
|
173
|
Lindsey ML, Bolli R, Canty JM, Du XJ, Frangogiannis NG, Frantz S, Gourdie RG, Holmes JW, Jones SP, Kloner RA, Lefer DJ, Liao R, Murphy E, Ping P, Przyklenk K, Recchia FA, Schwartz Longacre L, Ripplinger CM, Van Eyk JE, Heusch G. Guidelines for experimental models of myocardial ischemia and infarction. Am J Physiol Heart Circ Physiol 2018; 314:H812-H838. [PMID: 29351451 PMCID: PMC5966768 DOI: 10.1152/ajpheart.00335.2017] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is a prevalent major cardiovascular event that arises from myocardial ischemia with or without reperfusion, and basic and translational research is needed to better understand its underlying mechanisms and consequences for cardiac structure and function. Ischemia underlies a broad range of clinical scenarios ranging from angina to hibernation to permanent occlusion, and while reperfusion is mandatory for salvage from ischemic injury, reperfusion also inflicts injury on its own. In this consensus statement, we present recommendations for animal models of myocardial ischemia and infarction. With increasing awareness of the need for rigor and reproducibility in designing and performing scientific research to ensure validation of results, the goal of this review is to provide best practice information regarding myocardial ischemia-reperfusion and infarction models. Listen to this article’s corresponding podcast at ajpheart.podbean.com/e/guidelines-for-experimental-models-of-myocardial-ischemia-and-infarction/.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Research Service, G. V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Roberto Bolli
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
| | - John M Canty
- Division of Cardiovascular Medicine, Departments of Biomedical Engineering and Physiology and Biophysics, The Veterans Affairs Western New York Health Care System and Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, New York
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital , Würzburg , Germany
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia Health System , Charlottesville, Virginia
| | - Steven P Jones
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Robert A Kloner
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes , Pasadena, California.,Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana
| | - Ronglih Liao
- Harvard Medical School , Boston, Massachusetts.,Division of Genetics and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital , Boston, Massachusetts
| | - Elizabeth Murphy
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Peipei Ping
- National Institutes of Health BD2KBig Data to Knowledge (BD2K) Center of Excellence and Department of Physiology, Medicine and Bioinformatics, University of California , Los Angeles, California
| | - Karin Przyklenk
- Cardiovascular Research Institute and Departments of Physiology and Emergency Medicine, Wayne State University School of Medicine , Detroit, Michigan
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Fondazione G. Monasterio, Pisa , Italy.,Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University , Philadelphia, Pennsylvania
| | - Lisa Schwartz Longacre
- Heart Failure and Arrhythmias Branch, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California , Davis, California
| | - Jennifer E Van Eyk
- The Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center , Los Angeles, California
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School , Essen , Germany
| |
Collapse
|
174
|
Ren J, Li C, Liu Y, Liu H, Dong Z. Protective effect of dexmedetomidine against myocardial ischemia-reperfusion injury in rabbits. Acta Cir Bras 2018; 33:22-30. [DOI: 10.1590/s0102-865020180010000003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Affiliation(s)
| | - Changfa Li
- Zaozhuang Maternity and Child Care Centers, China
| | - Yan Liu
- Hebei Medical University, China
| | | | | |
Collapse
|
175
|
Affiliation(s)
- Robert A Kloner
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
176
|
Prognostic Association of Circulating Neutrophil Count with No-Reflow in Patients with ST-Segment Elevation Myocardial Infarction following Successful Primary Percutaneous Intervention. DISEASE MARKERS 2017; 2017:8458492. [PMID: 29379223 PMCID: PMC5742887 DOI: 10.1155/2017/8458492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/30/2017] [Accepted: 10/31/2017] [Indexed: 01/04/2023]
Abstract
Objective The aim of the present study was to investigate the predictive value of neutrophil count for no-reflow in patients with ST-segment elevation myocardial infarction (STEMI) who underwent successful primary percutaneous intervention (PCI). Methods We conducted a retrospective study of 361 patients diagnosed with acute STEMI between 2011 and 2015. All patients underwent successful PCI within 12 h from the onset of symptoms. Angiographic no-reflow was diagnosed based on a post-PCI thrombolysis in myocardial infarction flow grade ≤ 2 without mechanical obstruction. According to a neutrophil count cut-off determined by receiver operating characteristic curve analysis, patients were divided into two groups: group A (neutrophil count < 9.14 × 109/L) and group B (neutrophil count ≥ 9.14 × 109/L). Results Compared to patients in the normal reflow group, patients with no-reflow had higher neutrophil counts (P < 0.05). The incidence rate of no-reflow in group A (18, 9.3%) was significantly lower than that in group B (38). Multivariate logistic regression analysis revealed that a neutrophil count ≥ 9.14 × 109/L was independently predictive for no-reflow (odds ratio = 4.474, 95% confidence interval: 1.610–12.433, P = 0.004) after adjusting for potential confounders. Conclusions A circulating neutrophil count ≥ 9.14 × 109/L is independently associated with no-reflow in patients with acute STEMI following primary PCI.
Collapse
|
177
|
Davies JMS, Cillard J, Friguet B, Cadenas E, Cadet J, Cayce R, Fishmann A, Liao D, Bulteau AL, Derbré F, Rébillard A, Burstein S, Hirsch E, Kloner RA, Jakowec M, Petzinger G, Sauce D, Sennlaub F, Limon I, Ursini F, Maiorino M, Economides C, Pike CJ, Cohen P, Salvayre AN, Halliday MR, Lundquist AJ, Jakowec NA, Mechta-Grigoriou F, Mericskay M, Mariani J, Li Z, Huang D, Grant E, Forman HJ, Finch CE, Sun PY, Pomatto LCD, Agbulut O, Warburton D, Neri C, Rouis M, Cillard P, Capeau J, Rosenbaum J, Davies KJA. The Oxygen Paradox, the French Paradox, and age-related diseases. GeroScience 2017; 39:499-550. [PMID: 29270905 PMCID: PMC5745211 DOI: 10.1007/s11357-017-0002-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023] Open
Abstract
A paradox is a seemingly absurd or impossible concept, proposition, or theory that is often difficult to understand or explain, sometimes apparently self-contradictory, and yet ultimately correct or true. How is it possible, for example, that oxygen "a toxic environmental poison" could be also indispensable for life (Beckman and Ames Physiol Rev 78(2):547-81, 1998; Stadtman and Berlett Chem Res Toxicol 10(5):485-94, 1997)?: the so-called Oxygen Paradox (Davies and Ursini 1995; Davies Biochem Soc Symp 61:1-31, 1995). How can French people apparently disregard the rule that high dietary intakes of cholesterol and saturated fats (e.g., cheese and paté) will result in an early death from cardiovascular diseases (Renaud and de Lorgeril Lancet 339(8808):1523-6, 1992; Catalgol et al. Front Pharmacol 3:141, 2012; Eisenberg et al. Nat Med 22(12):1428-1438, 2016)?: the so-called, French Paradox. Doubtless, the truth is not a duality and epistemological bias probably generates apparently self-contradictory conclusions. Perhaps nowhere in biology are there so many apparently contradictory views, and even experimental results, affecting human physiology and pathology as in the fields of free radicals and oxidative stress, antioxidants, foods and drinks, and dietary recommendations; this is particularly true when issues such as disease-susceptibility or avoidance, "healthspan," "lifespan," and ageing are involved. Consider, for example, the apparently paradoxical observation that treatment with low doses of a substance that is toxic at high concentrations may actually induce transient adaptations that protect against a subsequent exposure to the same (or similar) toxin. This particular paradox is now mechanistically explained as "Adaptive Homeostasis" (Davies Mol Asp Med 49:1-7, 2016; Pomatto et al. 2017a; Lomeli et al. Clin Sci (Lond) 131(21):2573-2599, 2017; Pomatto and Davies 2017); the non-damaging process by which an apparent toxicant can activate biological signal transduction pathways to increase expression of protective genes, by mechanisms that are completely different from those by which the same agent induces toxicity at high concentrations. In this review, we explore the influences and effects of paradoxes such as the Oxygen Paradox and the French Paradox on the etiology, progression, and outcomes of many of the major human age-related diseases, as well as the basic biological phenomenon of ageing itself.
Collapse
Affiliation(s)
- Joanna M S Davies
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Josiane Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Bertrand Friguet
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Enrique Cadenas
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jean Cadet
- Département de Médecine nucléaire et Radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Rachael Cayce
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Andrew Fishmann
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - David Liao
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Anne-Laure Bulteau
- Institut de Génomique Fonctionnelle de Lyon,ENS de Lyon, CNRS, 69364, Lyon Cedex 07, France
| | - Frédéric Derbré
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Amélie Rébillard
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Steven Burstein
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Etienne Hirsch
- INSERM UMR 1127-CNRS UMR 7225, Institut du cerveau et de la moelle épinière-ICM Thérapeutique Expérimentale de la Maladie de Parkinson, Université Pierre et Marie Curie, 75651, Paris Cedex 13, France
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, 91105, USA
| | - Michael Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Giselle Petzinger
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Delphine Sauce
- Chronic infections and Immune ageing, INSERM U1135, Hopital Pitie-Salpetriere, Pierre et Marie Curie University, 75013, Paris, France
| | | | - Isabelle Limon
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Matilde Maiorino
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Christina Economides
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Christian J Pike
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Neurobiology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Anne Negre Salvayre
- Lipid peroxidation, Signalling and Vascular Diseases INSERM U1048, 31432, Toulouse Cedex 4, France
| | - Matthew R Halliday
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Adam J Lundquist
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicolaus A Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Mathias Mericskay
- Laboratoire de Signalisation et Physiopathologie Cardiovasculaire-Inserm UMR-S 1180, Faculté de Pharmacie, Université Paris-Sud, 92296 Châtenay-Malabry, Paris, France
| | - Jean Mariani
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Zhenlin Li
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - David Huang
- Department of Radiation Oncology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Ellsworth Grant
- Department of Oncology & Hematology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Henry J Forman
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Patrick Y Sun
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Laura C D Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Onnik Agbulut
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - David Warburton
- Children's Hospital of Los Angeles, Developmental Biology, Regenerative Medicine and Stem Cell Therapeutics program and the Center for Environmental Impact on Global Health Across the Lifespan at The Saban Research Institute, Los Angeles, CA, 90027, USA
- Department of Pediatrics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Christian Neri
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Mustapha Rouis
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Pierre Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Jacqueline Capeau
- DR Saint-Antoine UMR_S938, UPMC, Inserm Faculté de Médecine, Université Pierre et Marie Curie, 75012, Paris, France
| | - Jean Rosenbaum
- Scientific Service of the Embassy of France in the USA, Consulate General of France in Los Angeles, Los Angeles, CA, 90025, USA
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA.
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA.
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA.
| |
Collapse
|
178
|
O'Farrell FM, Mastitskaya S, Hammond-Haley M, Freitas F, Wah WR, Attwell D. Capillary pericytes mediate coronary no-reflow after myocardial ischaemia. eLife 2017; 6:29280. [PMID: 29120327 PMCID: PMC5705208 DOI: 10.7554/elife.29280] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/08/2017] [Indexed: 12/22/2022] Open
Abstract
After cardiac ischaemia, a prolonged decrease of coronary microvascular perfusion often occurs even after flow is restored in an upstream artery. This 'no-reflow' phenomenon worsens patient prognosis. In the brain, after stroke, a similar post-ischaemic 'no-reflow' has been attributed to capillary constriction by contractile pericytes. We now show that occlusion of a rat coronary artery, followed by reperfusion, blocks 40% of cardiac capillaries and halves perfused blood volume within the affected region. Capillary blockages colocalised strongly with pericytes, where capillary diameter was reduced by 37%. The pericyte relaxant adenosine increased capillary diameter by 21% at pericyte somata, decreased capillary block by 25% and increased perfusion volume by 57%. Thus, cardiac pericytes constrict coronary capillaries and reduce microvascular blood flow after ischaemia, despite re-opening of the culprit artery. Cardiac pericytes are therefore a novel therapeutic target in ischaemic heart disease. Heart attacks occur when one of the arteries supplying blood to the heart muscle becomes blocked, usually by a blood clot. Doctors unblock the artery and insert an expanding metal cage called a stent to keep it unblocked. This restores blood flow through the artery. Unfortunately, blood flow often does not return to smaller downstream blood vessels called capillaries. This can lead to further damage to the heart. Scientists have not been able to find a way to reliably open up those capillaries after a heart attack because it is not clear exactly what is keeping them closed. Muscle-like cells called pericytes, which wrap around the capillaries, are one possible culprit for the blockages. Pericytes narrow capillaries in the brain after stroke in animal experiments. These cells are also present on heart capillaries, but scientists do not know much about them. Now, O’Farrell, Mastitskaya, Hammond-Haley et al. show that pericytes are partly responsible for limiting blood flow in capillaries after a heart attack in rats. In the experiments, blood flow through an artery feeding the hearts of anaesthetized rats was restricted, simulating a heart attack. After the blood flow was later restored, 40% of the animal’s capillaries remained blocked. Many blockages occurred near pericytes that had narrowed the capillary preventing blood flow. Treating the rats with a drug called adenosine, which relaxes the pericytes, reduced capillary blockages and increased blood flow in the heart. Although adenosine could help to restore blood flow in the capillaries after a heart attack, it may also relax muscles around arteries and lower blood pressure, and so it may not be an ideal treatment. More studies are needed to determine whether drugs that target only the pericytes could complement existing heart attack treatments that unblock the arteries. If these studies are successful, pericyte-targeting drugs might prevent serious complications after a heart attack, including heart failure, heart rhythm abnormalities and future heart attacks.
Collapse
Affiliation(s)
- Fergus M O'Farrell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Svetlana Mastitskaya
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Matthew Hammond-Haley
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Felipe Freitas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Wen Rui Wah
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
179
|
Kloner RA, Brown DA, Csete M, Dai W, Downey JM, Gottlieb RA, Hale SL, Shi J. New and revisited approaches to preserving the reperfused myocardium. Nat Rev Cardiol 2017; 14:679-693. [PMID: 28748958 PMCID: PMC5991096 DOI: 10.1038/nrcardio.2017.102] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Early coronary artery reperfusion improves outcomes for patients with ST-segment elevation myocardial infarction (STEMI), but morbidity and mortality after STEMI remain unacceptably high. The primary deficits seen in these patients include inadequate pump function, owing to rapid infarction of muscle in the first few hours of treatment, and adverse remodelling of the heart in the months that follow. Given that attempts to further reduce myocardial infarct size beyond early reperfusion in clinical trials have so far been disappointing, effective therapies are still needed to protect the reperfused myocardium. In this Review, we discuss several approaches to preserving the reperfused heart, such as therapies that target the mechanisms involved in mitochondrial bioenergetics, pyroptosis, and autophagy, as well as treatments that harness the cardioprotective properties of inhaled anaesthetic agents. We also discuss potential therapies focused on correcting the no-reflow phenomenon and its effect on healing and adverse left ventricular remodelling.
Collapse
Affiliation(s)
- Robert A Kloner
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
- Virginia Tech Center for Drug Discovery, Virginia Tech, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
- Virginia Tech Metabolic Phenotyping Core, Virginia Tech, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Marie Csete
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Department of Anesthesiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90017, USA
| | - Wangde Dai
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| | - James M Downey
- Department of Physiology and Cell Biology, University of South Alabama, 5851 USA Drive North, Mobile, Alabama 36688, USA
| | - Roberta A Gottlieb
- Department of Medicine, Barbra Streisand Women's Heart Center, Heart Institute of Cedars-Sinai, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, California 90048, USA
| | - Sharon L Hale
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
| | - Jianru Shi
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| |
Collapse
|
180
|
Zhou H, Zhu P, Guo J, Hu N, Wang S, Li D, Hu S, Ren J, Cao F, Chen Y. Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol 2017; 13:498-507. [PMID: 28732308 PMCID: PMC5828768 DOI: 10.1016/j.redox.2017.07.007] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/03/2017] [Accepted: 07/08/2017] [Indexed: 02/06/2023] Open
Abstract
Ripk3-required necroptosis and mitochondria-mediated apoptosis are the predominant types of cell death that largely account for the development of cardiac ischemia reperfusion injury (IRI). Here, we explored the effect of Ripk3 on mitochondrial apoptosis. Compared with wild-type mice, the infarcted area in Ripk3-deficient (Ripk3-/-) mice had a relatively low abundance of apoptotic cells. Moreover, the loss of Ripk3 protected the mitochondria against IRI and inhibited caspase9 apoptotic pathways. These protective effects of Ripk3 deficiency were relied on mitophagy activation. However, inhibition of mitophagy under Ripk3 deficiency enhanced cardiomyocyte and endothelia apoptosis, augmented infarcted area and induced microvascular dysfunction. Furthermore, ischemia activated mitophagy by modifying FUNDC1 dephosphorylation, which substantively engulfed mitochondria debris and cytochrome-c, thus blocking apoptosis signal. However, reperfusion injury elevated the expression of Ripk3 which disrupted FUNDC1 activation and abated mitophagy, increasing the likelihood of apoptosis. In summary, this study confirms the promotive effect of Ripk3 on mitochondria-mediated apoptosis via inhibition of FUNDC1-dependent mitophagy in cardiac IRI. These findings provide new insight into the roles of Ripk3-related necroptosis, mitochondria-mediated apoptosis and FUNDC1-required mitophagy in cardiac IRI.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jun Guo
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Nan Hu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Shuyi Wang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Dandan Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
181
|
Toldo S, Austin D, Mauro AG, Mezzaroma E, Van Tassell BW, Marchetti C, Carbone S, Mogelsvang S, Gelber C, Abbate A. Low-Density Lipoprotein Receptor-Related Protein-1 Is a Therapeutic Target in Acute Myocardial Infarction. JACC Basic Transl Sci 2017; 2:561-574. [PMID: 30062170 PMCID: PMC6058925 DOI: 10.1016/j.jacbts.2017.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 01/29/2023]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is a ubiquitous membrane receptor functioning as a scavenger and regulatory receptor, inducing anti-inflammatory and prosurvival signals. Based on the known structure-activity of the LRP1 receptor binding site, the authors synthesized a small peptide (SP16). SP16 induced a >50% reduction in infarct size (p < 0.001) and preservation of left ventricular systolic function (p < 0.001), and treatment with an LRP1 blocking antibody eliminated the protective effects of SP16. In conclusion, LRP1 activation with SP16 given within 30 min of reperfusion during experimental acute myocardial infarction leads to a cardioprotective signal reducing infarct size and preservation of cardiac systolic function.
Collapse
Key Words
- A2MG, alpha-2 macroglobulin
- AAT, alpha-1 antitrypsin
- AMI, acute myocardial infarction
- ATIII, antithrombin III
- HRP, horseradish peroxidase
- IL, interleukin
- IV, intravenous
- LPS, lipopolysaccharide
- LRP1, low-density lipoprotein receptor–related protein-1
- LV, left ventricular
- LVFS, left ventricular fractional shortening
- PBS, phosphate-buffered saline
- SEC, serine protease inhibitor–enzyme complex
- SERPIN, serine protease inhibitor
- SERPINs
- TBS, tris-buffered saline
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- ischemia reperfusion
- low-density lipoprotein receptor-related protein-1
- serine protease inhibitor
Collapse
Affiliation(s)
- Stefano Toldo
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
| | | | - Adolfo G. Mauro
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
| | - Eleonora Mezzaroma
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Benjamin W. Van Tassell
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Carlo Marchetti
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
| | - Salvatore Carbone
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
| | | | | | - Antonio Abbate
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
- Johnson Research Center for Critical Care, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
182
|
Management of No-Reflow Phenomenon in the Catheterization Laboratory. JACC Cardiovasc Interv 2017; 10:215-223. [PMID: 28183461 DOI: 10.1016/j.jcin.2016.11.059] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/07/2016] [Accepted: 11/29/2016] [Indexed: 12/29/2022]
Abstract
At the conclusion of a primary percutaneous coronary intervention for ST-segment elevation myocardial infarction, and after the cardiologist makes certain that there is no residual stenosis following stenting, assessment of coronary flow becomes the top priority. The presence of no-reflow is a serious prognostic sign. No-reflow can result in poor healing of the infarct and adverse left ventricular remodeling, increasing the risk for major adverse cardiac events, including congestive heart failure and death. To achieve normal flow, features associated with a high incidence of no-reflow must be anticipated, and measures must be undertaken to prevent its occurrence. In this review, the authors discuss various preventive strategies for no-reflow as well as pharmacological and nonpharmacological interventions that improve coronary blood flow, such as intracoronary adenosine and nitroprusside. Nonpharmacological therapies, such as induced hypothermia, were successful in animal studies, but their effectiveness in reducing no-reflow in humans remains to be determined.
Collapse
|
183
|
Granger DN, Kvietys PR. Reperfusion therapy-What's with the obstructed, leaky and broken capillaries? ACTA ACUST UNITED AC 2017; 24:213-228. [PMID: 29102280 DOI: 10.1016/j.pathophys.2017.09.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Microvascular dysfunction is well established as an early and rate-determining factor in the injury response of tissues to ischemia and reperfusion (I/R). Severe endothelial cell dysfunction, which can develop without obvious morphological cell injury, is a major underlying cause of the microvascular abnormalities that accompany I/R. While I/R-induced microvascular dysfunction is manifested in different ways, two responses that have received much attention in both the experimental and clinical setting are impaired capillary perfusion (no-reflow) and endothelial barrier failure with a transition to hemorrhage. These responses are emerging as potentially important determinants of the severity of the tissue injury response, and there is growing clinical evidence that they are predictive of clinical outcome following reperfusion therapy. This review provides a summary of animal studies that have focused on the mechanisms that may underlie the genesis of no-reflow and hemorrhage following reperfusion of ischemic tissues, and addresses the clinical evidence that implicates these vascular events in the responses of the ischemic brain (stroke) and heart (myocardial infarction) to reperfusion therapy. Inasmuch as reactive oxygen species (ROS) and matrix metalloproteinases (MMP) are frequently invoked as triggers of the microvascular dysfunction elicited by I/R, the potential roles and sources of these mediators are also discussed. The available evidence in the literature justifies the increased interest in the development of no-reflow and hemorrhage in heart and brain following reperfusion therapy, and suggests that these vascular events may be predictive of poor clinical outcome and warrant the development of targeted treatment strategies.
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
184
|
The Effect of Coenzyme Q10 (Ubiquinone) on Random Pattern Skin Flap Survival in Rat Model. Ann Plast Surg 2017; 77:e9-e14. [PMID: 25954836 DOI: 10.1097/sap.0000000000000504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In this study, the effect of coenzyme Q10 (CQ10) on flap survival was investigated. METHODS Fifty Wistar Albino rats were divided into 5 groups. The survival rates of the skin flaps were assessed 10 days after complete elevation of the flaps. Regions of survival and necrosis were drawn on transparent acetate sheets and scanned into a computer. Tissue samples were assessed histopathologically after staining with hematoxylin-eosin, vascular endothelial growth factor staining and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-Biotin Nick End-labeling staining. To evaluate the antioxidant effect of CQ10; malondialdehyde, nitric oxide levels were measured. RESULTS Viable flaps area was found higher in groups 3 and 4 as compared to groups 1, 2, and 5. In terms of vascular proliferation, elevated angiogenesis was observed in pathological specimens of groups 3 and 4 as compared to groups 1, 2, and 5. Malondialdehyde levels in groups 3 and 4 were found to be significantly decreased as compared to groups 1, 2 and 5 (P < 0.05). Moreover, serum levels of CQ10 were found significantly increased in groups 3 and 4 (P < 0.05). CONCLUSIONS In conclusion, CQ10 significantly improves flap viability in rat model, and the highest levels of serum CQ10 can be obtained by oral administration.
Collapse
|
185
|
Abstract
PURPOSE OF REVIEW The purpose of the present paper is to analytically review the diagnostic and prognostic role of CMR in ST-segment elevation myocardial infarction (STEMI) survivors. Percutaneous coronary intervention (PCI) is the treatment of choice in patients STEMI. However, risk of future events remains substantial. Assessment of the extent of myocardial infarction (MI), cardiac function and ventricular remodelling has become the focus of recent studies. Electrocardiography, angiography and echocardiography parameters, as well as risk scores, lack sensitivity and reproducibility in predicting future cardiovascular events. A major advantage of cardiac magnetic resonance imaging (CMR) is that it provides myocardial tissue characterization. RECENT FINDINGS CMR is able to quantify both reversible and irreversible myocardial injury and correlates with future events. This review will illustrate how microvascular function indices (myocardial salvage index, presence and amount of microvascular obstruction and intramyocardial haemorrhage) detectable by CMR add prognostic information and could impact on future strategies to improve outcomes in revascularized patients.
Collapse
|
186
|
Skyschally A, Amanakis G, Neuhäuser M, Kleinbongard P, Heusch G. Impact of electrical defibrillation on infarct size and no-reflow in pigs subjected to myocardial ischemia-reperfusion without and with ischemic conditioning. Am J Physiol Heart Circ Physiol 2017; 313:H871-H878. [PMID: 28778913 DOI: 10.1152/ajpheart.00293.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/16/2022]
Abstract
Ventricular fibrillation (VF) occurs frequently during myocardial ischemia-reperfusion (I/R) and must then be terminated by electrical defibrillation. We have investigated the impact of VF/defibrillation on infarct size (IS) or area of no reflow (NR) without and with ischemic conditioning interventions. Anesthetized pigs were subjected to 60/180 min of coronary occlusion/reperfusion. VF, as identified from the ECG, was terminated by intrathoracic defibrillation. The area at risk (AAR), IS, and NR were determined by staining techniques (patent blue, triphenyltetrazolium chloride, and thioflavin-S). Four experimental protocols were analyzed: I/R (n = 49), I/R with ischemic preconditioning (IPC; n = 22), I/R with ischemic postconditioning (POCO; n = 22), or I/R with remote IPC (RIPC; n = 34). The incidence of VF was not different between I/R (44%), IPC (45%), POCO (50%), and RIPC (33%). IS was reduced by IPC (23 ± 12% of AAR), POCO (31 ± 16%), and RIPC (22 ± 13%, all P < 0.05 vs. I/R: 41 ± 12%). NR was not different between protocols (I/R: 17 ± 15% of AAR, IPC: 15 ± 18%, POCO: 25 ± 16%, and RIPC: 18 ± 17%). In pigs with defibrillation, IS was 50% larger than in pigs without defibrillation but independent of the number of defibrillations. Analysis of covariance confirmed the established determinants of IS, i.e., AAR, residual blood flow during ischemia (RMBFi), and a conditioning protocol, and revealed VF/defibrillation as a novel covariate. VF/defibrillation in turn was associated with larger AAR and lower RMBFi. Lack of dose-response relation between IS and the number of defibrillations excluded direct electrical injury as the cause of increased IS. Obviously, AAR size and RMBFi account for both IS and the incidence of VF. IS and NR are mechanistically distinct phenomena.NEW & NOTEWORTHY Ventricular fibrillation/defibrillation is associated with increased infarct size. Electrical injury is unlikely the cause of such association, since there is no dose-response relation between infarct size and number of defibrillations. Ventricular fibrillation, in turn, is associated with a larger area at risk and lower residual blood flow.
Collapse
Affiliation(s)
- Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany; and
| | - Georgios Amanakis
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany; and
| | - Markus Neuhäuser
- Department of Mathematics and Technology, Koblenz University of Applied Sciences, Rhein-Ahr-Campus, Remagen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany; and
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany; and
| |
Collapse
|
187
|
Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F, Chen Y. Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res 2017; 63:e12413. [PMID: 28398674 PMCID: PMC5518188 DOI: 10.1111/jpi.12413] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/07/2017] [Indexed: 12/18/2022]
Abstract
The cardiac microvascular system, which is primarily composed of monolayer endothelial cells, is the site of blood supply and nutrient exchange to cardiomyocytes. However, microvascular ischemia/reperfusion injury (IRI) following percutaneous coronary intervention is a woefully neglected topic, and few strategies are available to reverse such pathologies. Here, we studied the effects of melatonin on microcirculation IRI and elucidated the underlying mechanism. Melatonin markedly reduced infarcted area, improved cardiac function, restored blood flow, and lower microcirculation perfusion defects. Histological analysis showed that cardiac microcirculation endothelial cells (CMEC) in melatonin-treated mice had an unbroken endothelial barrier, increased endothelial nitric oxide synthase expression, unobstructed lumen, reduced inflammatory cell infiltration, and less endothelial damage. In contrast, AMP-activated protein kinase α (AMPKα) deficiency abolished the beneficial effects of melatonin on microvasculature. In vitro, IRI activated dynamin-related protein 1 (Drp1)-dependent mitochondrial fission, which subsequently induced voltage-dependent anion channel 1 (VDAC1) oligomerization, hexokinase 2 (HK2) liberation, mitochondrial permeability transition pore (mPTP) opening, PINK1/Parkin upregulation, and ultimately mitophagy-mediated CMEC death. However, melatonin strengthened CMEC survival via activation of AMPKα, followed by p-Drp1S616 downregulation and p-Drp1S37 upregulation, which blunted Drp1-dependent mitochondrial fission. Suppression of mitochondrial fission by melatonin recovered VDAC1-HK2 interaction that prevented mPTP opening and PINK1/Parkin activation, eventually blocking mitophagy-mediated cellular death. In summary, this study confirmed that melatonin protects cardiac microvasculature against IRI. The underlying mechanism may be attributed to the inhibitory effects of melatonin on mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis via activation of AMPKα.
Collapse
Affiliation(s)
- Hao Zhou
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Ying Zhang
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Shunying Hu
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Chen Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Radiation OncologyPeking University Cancer Hospital and InstituteBeijingChina
| | - Pingjun Zhu
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Qiang Ma
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Qinhua Jin
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Feng Cao
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Feng Tian
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Yundai Chen
- Department of CardiologyChinese PLA General HospitalBeijingChina
| |
Collapse
|
188
|
Liu D, Borlotti A, Viliani D, Jerosch-Herold M, Alkhalil M, De Maria GL, Fahrni G, Dawkins S, Wijesurendra R, Francis J, Ferreira V, Piechnik S, Robson MD, Banning A, Choudhury R, Neubauer S, Channon K, Kharbanda R, Dall'Armellina E. CMR Native T1 Mapping Allows Differentiation of Reversible Versus Irreversible Myocardial Damage in ST-Segment-Elevation Myocardial Infarction: An OxAMI Study (Oxford Acute Myocardial Infarction). Circ Cardiovasc Imaging 2017; 10:e005986. [PMID: 28798137 PMCID: PMC5555391 DOI: 10.1161/circimaging.116.005986] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 07/12/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND CMR T1 mapping is a quantitative imaging technique allowing the assessment of myocardial injury early after ST-segment-elevation myocardial infarction. We sought to investigate the ability of acute native T1 mapping to differentiate reversible and irreversible myocardial injury and its predictive value for left ventricular remodeling. METHODS AND RESULTS Sixty ST-segment-elevation myocardial infarction patients underwent acute and 6-month 3T CMR, including cine, T2-weighted (T2W) imaging, native shortened modified look-locker inversion recovery T1 mapping, rest first pass perfusion, and late gadolinium enhancement. T1 cutoff values for oedematous versus necrotic myocardium were identified as 1251 ms and 1400 ms, respectively, with prediction accuracy of 96.7% (95% confidence interval, 82.8% to 99.9%). Using the proposed threshold of 1400 ms, the volume of irreversibly damaged tissue was in good agreement with the 6-month late gadolinium enhancement volume (r=0.99) and correlated strongly with the log area under the curve troponin (r=0.80) and strongly with 6-month ejection fraction (r=-0.73). Acute T1 values were a strong predictor of 6-month wall thickening compared with late gadolinium enhancement. CONCLUSIONS Acute native shortened modified look-locker inversion recovery T1 mapping differentiates reversible and irreversible myocardial injury, and it is a strong predictor of left ventricular remodeling in ST-segment-elevation myocardial infarction. A single CMR acquisition of native T1 mapping could potentially represent a fast, safe, and accurate method for early stratification of acute patients in need of more aggressive treatment. Further confirmatory studies will be needed.
Collapse
Affiliation(s)
- Dan Liu
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Alessandra Borlotti
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Dafne Viliani
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Michael Jerosch-Herold
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Mohammad Alkhalil
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Giovanni Luigi De Maria
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Gregor Fahrni
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Sam Dawkins
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Rohan Wijesurendra
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Jane Francis
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Vanessa Ferreira
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Stefan Piechnik
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Matthew D Robson
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Adrian Banning
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Robin Choudhury
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Stefan Neubauer
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Keith Channon
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Rajesh Kharbanda
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.)
| | - Erica Dall'Armellina
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, United Kingdom (D.L., A.B., D.V., M.A., G.L.D.M., G.F., S.D., R.W., J.F., V.F., S.P., M.D.R., R.C., S.N., E.D.A.); Department of Cardiovascular Medicine, Oxford Heart Centre, John Radcliffe Hospital, Headley Way, United Kingdom (A.B., K.C., R.K.); and Department of Radiology, Brigham and Women's Hospital, Boston, MA (M.J.-H.).
| |
Collapse
|
189
|
Ghugre NR, Pop M, Thomas R, Newbigging S, Qi X, Barry J, Strauss BH, Wright GA. Hemorrhage promotes inflammation and myocardial damage following acute myocardial infarction: insights from a novel preclinical model and cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2017; 19:50. [PMID: 28676061 PMCID: PMC5496389 DOI: 10.1186/s12968-017-0361-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/09/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Myocardial hemorrhage is a frequent complication following reperfusion in acute myocardial infarction and is predictive of adverse outcomes. However, it remains unsettled whether hemorrhage is simply a marker of a severe initial ischemic insult or directly contributes to downstream myocardial damage. Our objective was to evaluate the contribution of hemorrhage towards inflammation, microvascular obstruction and infarct size in a novel porcine model of hemorrhagic myocardial infarction using cardiovascular magnetic resonance (CMR). METHODS Myocardial hemorrhage was induced via direct intracoronary injection of collagenase in a novel porcine model of ischemic injury. Animals (N = 27) were subjected to coronary balloon occlusion followed by reperfusion and divided into three groups (N = 9/group): 8 min ischemia with collagenase (+HEM); 45 min infarction with saline (I-HEM); and 45 min infarction with collagenase (I+HEM). Comprehensive CMR was performed on a 3 T scanner at baseline and 24 h post-intervention. Cardiac function was quantified by cine imaging, edema/inflammation by T2 mapping, hemorrhage by T2* mapping and infarct/microvascular obstruction size by gadolinium enhancement. Animals were subsequently sacrificed and explanted hearts underwent histopathological assessment for ischemic damage and inflammation. RESULTS At 24 h, the +HEM group induced only hemorrhage, the I-HEM group resulted in a non-hemorrhagic infarction, and the I+HEM group resulted in infarction and hemorrhage. Notably, the I+HEM group demonstrated greater hemorrhage and edema, larger infarct size and higher incidence of microvascular obstruction. Interestingly, hemorrhage alone (+HEM) also resulted in an observable inflammatory response, similar to that arising from a mild ischemic insult (I-HEM). CMR findings were in good agreement with histological staining patterns. CONCLUSIONS Hemorrhage is not simply a bystander, but an active modulator of tissue response, including inflammation and microvascular and myocardial damage beyond the initial ischemic insult. A mechanistic understanding of the pathophysiology of reperfusion hemorrhage will potentially aid better management of high-risk patients who are prone to adverse long-term outcomes.
Collapse
Affiliation(s)
- Nilesh R. Ghugre
- Physical Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Room M7-510, Toronto, ON M4N 3M5 Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Schulich Heart Research Program, Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | - Mihaela Pop
- Physical Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Room M7-510, Toronto, ON M4N 3M5 Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Schulich Heart Research Program, Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | - Reuben Thomas
- Physical Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Room M7-510, Toronto, ON M4N 3M5 Canada
| | - Susan Newbigging
- The Toronto Centre for Phenogenomics, Mount Sinai Hospital, Toronto, ON Canada
| | - Xiuling Qi
- Physical Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Room M7-510, Toronto, ON M4N 3M5 Canada
| | - Jennifer Barry
- Physical Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Room M7-510, Toronto, ON M4N 3M5 Canada
| | - Bradley H. Strauss
- Schulich Heart Research Program, Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | - Graham A. Wright
- Physical Sciences Platform, Sunnybrook Research Institute, 2075 Bayview Avenue, Room M7-510, Toronto, ON M4N 3M5 Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Schulich Heart Research Program, Sunnybrook Health Sciences Centre, Toronto, ON Canada
| |
Collapse
|
190
|
Elgavish GA, Simor T, van der Geest RJ, Suranyi P, Kiss PP, Lenkey Z, Kirschner R, Wang D, Brott BC, Varga-Szemes A. The MRI characteristics of the no-flow region are similar in reperfused and non-reperfused myocardial infarcts: an MRI and histopathology study in swine. Eur Radiol Exp 2017; 1:2. [PMID: 29708171 PMCID: PMC5909333 DOI: 10.1186/s41747-017-0001-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/08/2017] [Indexed: 11/10/2022] Open
Abstract
Background The no-flow region (NF) visualised by magnetic resonance imaging (MRI) in myocardial infarction (MI) has been explained as the product of reperfusion-injury-induced microvascular obstruction. However, a similar MRI phenomenon occurs in non-reperfused MI. Accordingly, our purpose was to compare the MRI and histopathologic characteristics of the NF in reperfused and non-reperfused MIs. Methods Reperfused (n = 7) and non-reperfused MIs (n = 7) were generated in swine by percutaneous balloon occlusion and microsphere embolisation techniques. Four days post-MI, animals underwent myocardial T2-mapping, early and serial late gadolinium enhancement MRI. MI and NF were compared between the models using the independent samples t test. Serial measurements were analysed using repeated measures analysis of variance. Triphenyltetrazolium chloride (TTC) macroscopic and microscopic histopathologic assessment was also performed. Results The MI size in the reperfused and non-reperfused groups was 17.1 ± 3.4 ml and 19.4 ± 8.1 ml, respectively (p = 0.090), in agreement with TTC assessment (p = 0.216; p = 0.484), and the NF size was 7.7 ± 2.4 ml and 8.1 ± 1.9 ml, respectively (P = 0.211). Compared to the reference 2-min post-contrast measurement, the NF size was significantly reduced at 20 min in the reperfused group and at 25 min in the non-reperfused group (both p < 0.001). Nevertheless, the NF was still detectable at 45 min after injection. No significant T2 difference was observed between the groups (p > 0.326). Histopathologic assessment revealed extensive calcification and hemosiderin deposition in the NF of the reperfused MI, but not in the non-reperfused MI. Conclusions The NF in non-reperfused and reperfused MIs have similar characteristics on MRI despite the different pathophysiologic and underlying histopathologic conditions, indicating that the presence of the NF alone cannot differentiate between these two types of MI.
Collapse
Affiliation(s)
- Gabriel A Elgavish
- 1Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, MCLM 556, Birmingham, AL 35294-0005 USA.,2Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, FOT 907, Birmingham, AL 35294-3407 USA.,Elgavish Paramagnetics Inc., 1737 Valpar Dr, Hoover, AL 35226 USA
| | - Tamas Simor
- 1Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, MCLM 556, Birmingham, AL 35294-0005 USA.,Elgavish Paramagnetics Inc., 1737 Valpar Dr, Hoover, AL 35226 USA.,4Heart Institute, Medical School, University of Pecs, Szigeti ut 12, Pecs, 7624 Hungary
| | - Rob J van der Geest
- 5Department of Radiology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA The Netherlands
| | - Pal Suranyi
- 6Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, 25 Courtenay Dr, Charleston, SC 29425 USA
| | - Pal P Kiss
- Elgavish Paramagnetics Inc., 1737 Valpar Dr, Hoover, AL 35226 USA
| | - Zsofia Lenkey
- 1Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, MCLM 556, Birmingham, AL 35294-0005 USA.,Elgavish Paramagnetics Inc., 1737 Valpar Dr, Hoover, AL 35226 USA.,4Heart Institute, Medical School, University of Pecs, Szigeti ut 12, Pecs, 7624 Hungary
| | - Robert Kirschner
- 1Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, MCLM 556, Birmingham, AL 35294-0005 USA.,Elgavish Paramagnetics Inc., 1737 Valpar Dr, Hoover, AL 35226 USA
| | - Dezhi Wang
- 7Department of Pathology, Histomorphometry and Molecular Analysis Core, University of Alabama at Birmingham, LHRB 589A, Birmingham, AL 35294-0007 USA
| | - Brigitta C Brott
- 2Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, FOT 907, Birmingham, AL 35294-3407 USA
| | - Akos Varga-Szemes
- 1Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, MCLM 556, Birmingham, AL 35294-0005 USA.,6Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, 25 Courtenay Dr, Charleston, SC 29425 USA
| |
Collapse
|
191
|
Koyama T. Lactated Ringer's solution for preventing myocardial reperfusion injury. IJC HEART & VASCULATURE 2017; 15:1-8. [PMID: 28616565 PMCID: PMC5458128 DOI: 10.1016/j.ijcha.2017.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/31/2017] [Indexed: 11/30/2022]
Abstract
Reperfusion of ischemic myocardium is crucial for salvaging myocardial cells from ischemic cell death. However, reperfusion itself induces various deleterious effects on the ischemic myocardium. These effects, known collectively as reperfusion injury, comprise stunned myocardium, reperfusion-induced arrhythmia, microvascular reperfusion injury, and lethal reperfusion injury. No approach has proven successful in preventing any of these injuries in the clinical setting. My colleagues and I recently proposed a new postconditioning protocol, postconditioning with lactate-enriched blood (PCLeB), for the prevention of reperfusion injury. This new approach consists of intermittent reperfusion and timely coronary injections of lactated Ringer's solution, aiming to achieve controlled reperfusion with cellular oxygenation and minimal lactate washout from the cells. This approach appeared to be effective in preventing all types of reperfusion injury in patients with ST-segment elevation myocardial infarction (STEMI), and we have already reported excellent in-hospital outcomes of patients with STEMI treated using PCLeB. In this review article, I discuss a possible mechanism of reperfusion injury, which we believe to be valid and which we targeted using this new approach, and I report how the approach worked in preventing each type of reperfusion injury.
Collapse
Key Words
- CAG, coronary angiography
- CK, creatine kinase
- CRP, C-reactive protein
- ECG, electrocardiography
- Lactate
- MI, myocardial infarction
- MPT, mitochondrial permeability transition
- No-reflow phenomenon
- PCI, percutaneous coronary intervention
- PCLeB, postconditioning with lactate-enriched blood
- PVC, premature ventricular contraction
- Postconditioning
- Reperfusion arrhythmia
- ST-segment elevation myocardial infarction
- STEMI, ST-segment elevation myocardial infarction
- Stunning
- TIMI, thrombolysis in myocardial infarction
- VF, ventricular fibrillation
- VT, ventricular tachycardia
Collapse
Affiliation(s)
- Takashi Koyama
- Cardiology Department, Saitama Municipal Hospital, 2460 Mimuro, Midori-ku, Saitama City, Saitama 336-8522, Japan
| |
Collapse
|
192
|
Towards accurate and precise T 1 and extracellular volume mapping in the myocardium: a guide to current pitfalls and their solutions. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2017; 31:143-163. [PMID: 28608328 PMCID: PMC5813078 DOI: 10.1007/s10334-017-0631-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/05/2017] [Accepted: 05/24/2017] [Indexed: 01/27/2023]
Abstract
Mapping of the longitudinal relaxation time (T1) and extracellular volume (ECV) offers a means of identifying pathological changes in myocardial tissue, including diffuse changes that may be invisible to existing T1-weighted methods. This technique has recently shown strong clinical utility for pathologies such as Anderson-Fabry disease and amyloidosis and has generated clinical interest as a possible means of detecting small changes in diffuse fibrosis; however, scatter in T1 and ECV estimates offers challenges for detecting these changes, and bias limits comparisons between sites and vendors. There are several technical and physiological pitfalls that influence the accuracy (bias) and precision (repeatability) of T1 and ECV mapping methods. The goal of this review is to describe the most significant of these, and detail current solutions, in order to aid scientists and clinicians to maximise the utility of T1 mapping in their clinical or research setting. A detailed summary of technical and physiological factors, issues relating to contrast agents, and specific disease-related issues is provided, along with some considerations on the future directions of the field.
Collapse
|
193
|
Zhang R, Chen S, Zhao Q, Sun M, Yu B, Hou J. Fragmented QRS complex is a prognostic marker of microvascular reperfusion and changes in LV function occur in patients with ST elevation myocardial infarction who underwent primary percutaneous coronary intervention. Exp Ther Med 2017; 13:3231-3238. [PMID: 28588675 PMCID: PMC5450773 DOI: 10.3892/etm.2017.4380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 01/20/2017] [Indexed: 01/08/2023] Open
Abstract
The present study aimed to investigate the in-hospital and long-term prognostic value of fragmented QRS complex (fQRS) for microvascular reperfusion and changes in left ventricular (LV) function in patients with ST elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PCI). A total of 216 patients with STEMI undergoing primary PCI were included in the current study. Patients were divided into two groups based on the presence (n=126) or absence (n=90) of fQRS following electrocardiograms (ECGs) on admission. Following primary PCI and follow up, patients were divided into four groups based on new onset, resolution, persistence and absence of fQRS. Major adverse cardiac events were defined to include cardiovascular death, arrhythmia, heart failure, reinfarction and target vessel revascularization. The percentage of patients with heart failure and microvascular reperfusion differed significantly between the fQRS(+) and fQRS(−) groups. Levels of N-terminal pro-brain natriuretic peptide (NT-proBNP), Peak creatine kinase-MB (CK-MB) and Troponin I levels were observed to be significantly higher in the fQRS(+) group compared with the fQRS(−) group. In univariate logistic regression analysis, left ventricular ejection fraction (LVEF), NT-proBNP, Troponin I, Peak CK-MB and microvascular reperfusion were found to be associated with fQRS. Multivariate analysis identified that LVEF, NT-proBNP, Troponin I and microvascular reperfusion may be independent predictors of fQRS. The presence of fQRS was demonstrated to be associated with left ventricular dysfunction at follow up assessments. The presence of fQRS was not only significantly associated with myocardial microvascular reperfusion and left ventricular function, but was also a prognostic marker in STEMI.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Shuyuan Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Qi Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Meng Sun
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Jingbo Hou
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
194
|
Montone RA, Niccoli G, Minelli S, Fracassi F, Vetrugno V, Aurigemma C, Burzotta F, Porto I, Trani C, Crea F. Clinical outcome and correlates of coronary microvascular obstruction in latecomers after acute myocardial infarction. Int J Cardiol 2017; 236:30-35. [DOI: 10.1016/j.ijcard.2017.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/03/2017] [Indexed: 12/20/2022]
|
195
|
Saremi F. Cardiac MR Imaging in Acute Coronary Syndrome: Application and Image Interpretation. Radiology 2017; 282:17-32. [PMID: 28005512 DOI: 10.1148/radiol.2016152849] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute coronary syndrome (ACS) is a frequent cause of hospitalization and coronary interventions. Cardiac magnetic resonance (MR) imaging is an increasingly used technique for initial work-up of chest pain and early post-reperfusion and follow-up evaluation of ACS to identify patients at high risk of further cardiac events. Cardiac MR imaging can evaluate with accuracy a variety of prognostic indicators of myocardial damage, including regional myocardial dysfunction, infarct distribution, infarct size, myocardium at risk, microvascular obstruction, and intramyocardial hemorrhage in both acute setting and later follow-up examinations. In addition, MR imaging is useful to rule out other causes of acute chest pain in patients admitted to the emergency department. In this article, a brief explanation of the pathophysiology, classification, and treatment options for patients with ACS will be introduced. Indications of cardiac MR imaging in ACS patients will be reviewed and specific cardiac MR protocol, image interpretation, and potential diagnostic pitfalls will be discussed. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Farhood Saremi
- From the Department of Radiology, University of Southern California, USC University Hospital, 1500 San Pablo St, Los Angeles CA 90033
| |
Collapse
|
196
|
Fineschi M, Verna E, Mezzapelle G, Bartolini D, Turiano G, Manari A, Lucarelli K, Uguccioni L, Repetto A, Tarantini G. Assessing MICRO-vascular resistances via IMR to predict outcome in STEMI patients with multivessel disease undergoing primary PCI (AMICRO): Rationale and design of a prospective multicenter clinical trial. Am Heart J 2017; 187:37-44. [PMID: 28454806 DOI: 10.1016/j.ahj.2017.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/05/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND In STEMI patients treated with primary percutaneous coronary angioplasty (PPCI) the evaluation of coronary microcirculatory resistance index (IMR) predict the extent of microvascular damage and left ventricular (LV) remodeling. However, the impact of IMR on the clinical outcome after PPCI in patients with multivessel disease (MVD) remains unsettled. AIM We designed a prospective multicenter controlled clinical trial to evaluate the prognostic value of IMR in terms of clinical outcome and left ventricular remodeling in STEMI patients with MVD undergoing PPCI. METHODS AND DESIGN The study will involve 242 patients with MVD defines as the presence of at least a non-culprit lesion of >50% stenosis at index coronary angiography. Both fractional flow reserve (FFR) and IMR will be measured in the infarct-related artery (IRA) after successful PPCI. Measurements of FFR and IMR will be repeated in the IRA and performed in the non-culprit vessels at staged angiography. The non-culprit vessel lesions will be treated only in the presence of a FFR<0.75. A 2D echocardiographic evaluation of the left ventricular (LV) volumes and ejection fraction will be performed before hospital discharge and at 1-year follow-up. The primary end-point of the study will be the composite of cardiovascular death, re-hospitalization for heart failure and resuscitation or appropriate ICD shock during 1-year of follow-up. Secondary end-points will be the impact of IMR in predicting LV remodeling during follow-up and correlations between IMR and ST-segment resolution. Other secondary endpoints will be need for new revascularization, stent thrombosis and re-infarction of the non-culprit vessels territory. IMPLICATIONS If IMR significantly correlates with differences in outcome and LV remodeling, it will emerge as a potential prognostic index after PPCI in patients with MVD.
Collapse
Affiliation(s)
| | - Edoardo Verna
- Ospedale di Circolo e Fondazione Macchi, Università dell'Insubria, Varese (VA).
| | | | | | | | | | - Katia Lucarelli
- Ospedale Generale Regionale F. Miulli, Acquaviva delle Fonti (BA)
| | | | | | | |
Collapse
|
197
|
Abstract
The atherosclerotic coronary vasculature is not only the culprit but also a victim of myocardial ischemia/reperfusion injury. Manifestations of such injury are increased vascular permeability and edema, endothelial dysfunction and impaired vasomotion, microembolization of atherothrombotic debris, stasis with intravascular cell aggregates, and finally, in its most severe form, capillary destruction with hemorrhage. In animal experiments, local and remote ischemic pre- and postconditioning not only reduce infarct size but also these manifestations of coronary vascular injury, as do drugs which recruit signal transduction steps of conditioning. Clinically, no-reflow is frequently seen after interventional reperfusion, and it carries an adverse prognosis. The translation of cardioprotective interventions to clinical practice has been difficult to date. Only 4 drugs (brain natriuretic peptide, exenatide, metoprolol, and esmolol) stand unchallenged to date in reducing infarct size in patients with reperfused acute myocardial infarction; unfortunately, for these drugs, no information on their impact on the ischemic/reperfused coronary circulation is available.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Essen, Essen, Germany.
| |
Collapse
|
198
|
Boag SE, Andreano E, Spyridopoulos I. Lymphocyte Communication in Myocardial Ischemia/Reperfusion Injury. Antioxid Redox Signal 2017; 26:660-675. [PMID: 28006953 DOI: 10.1089/ars.2016.6940] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Myocardial ischemia/reperfusion (I/R) is an important complication of reperfusion therapy for myocardial infarction (MI). It is a complex process involving metabolic and immunological factors. To date, no effective treatment has been identified. Recent Advances: Previous research has focused on the role of innate immune cells in I/R injury. In recent years, increasing evidence has accumulated for an important role for adaptive immune cells, particularly T lymphocytes. Data from ST elevation MI patients have identified prognostic significance for lymphocyte counts, particularly postreperfusion lymphopenia. Dynamic changes in circulating CD4+ T cell subsets occurring early after reperfusion are associated with development of I/R injury in the form of microvascular obstruction. Transcoronary gradients in cell counts suggest sequestration of these cells into the reperfused myocardium. These findings support existing data from mouse models indicating a role for CD4+ T cells in I/R injury. It is clear, however, the effects of lymphocytes in the ischemic myocardium are time and subset specific, with some having protective effects, while others are pathogenic. CRITICAL ISSUES An understanding of the cellular events that lead to accumulation of lymphocytes in the myocardium, and their actions once there, is key to manipulating this process. Chemokines produced in response to ischemia and cellular injury have an important role, while lymphocyte-derived cytokines are critical in the balance between inflammation and healing. FUTURE DIRECTIONS Further research into the involvement of lymphocytes in myocardial I/R injury may allow development of targeted therapies, opening a new avenue of considerable therapeutic potential. Antioxid. Redox Signal. 26, 660-675.
Collapse
Affiliation(s)
- Stephen E Boag
- 1 Institute of Genetic Medicine, Newcastle University , Newcastle upon Tyne, United Kingdom .,2 Regional Department of Clinical Immunology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Emanuele Andreano
- 1 Institute of Genetic Medicine, Newcastle University , Newcastle upon Tyne, United Kingdom
| | - Ioakim Spyridopoulos
- 1 Institute of Genetic Medicine, Newcastle University , Newcastle upon Tyne, United Kingdom
| |
Collapse
|
199
|
Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, Ma Q, Tian F, Chen Y. Mff-Dependent Mitochondrial Fission Contributes to the Pathogenesis of Cardiac Microvasculature Ischemia/Reperfusion Injury via Induction of mROS-Mediated Cardiolipin Oxidation and HK2/VDAC1 Disassociation-Involved mPTP Opening. J Am Heart Assoc 2017; 6:JAHA.116.005328. [PMID: 28288978 PMCID: PMC5524036 DOI: 10.1161/jaha.116.005328] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background The cardiac microvascular system ischemia/reperfusion injury following percutaneous coronary intervention is a clinical thorny problem. This study explores the mechanisms by which ischemia/reperfusion injury induces cardiac microcirculation collapse. Methods and Results In wild‐type mice, mitochondrial fission factor (Mff) expression increased in response to acute microvascular ischemia/reperfusion injury. Compared with wild‐type mice, homozygous Mff‐deficient (Mffgt) mice exhibited a smaller infarcted area, restored cardiac function, improved blood flow, and reduced microcirculation perfusion defects. Histopathology analysis demonstrated that cardiac microcirculation endothelial cells (CMECs) in Mffgt mice had an intact endothelial barrier, recovered phospho‐endothelial nitric oxide synthase production, opened lumen, undivided mitochondrial structures, and less CMEC death. In vitro, Mff‐deficient CMECs (derived from Mffgt mice or Mff small interfering RNA–treated) demonstrated less mitochondrial fission and mitochondrial‐dependent apoptosis compared with cells derived from wild‐type mice. The loss of Mff inhibited mitochondrial permeability transition pore opening via blocking the oligomerization of voltage‐dependent anion channel 1 and subsequent hexokinase 2 separation from mitochondria. Moreover, Mff deficiency reduced the cyt‐c leakage into the cytoplasm by alleviating cardiolipin oxidation resulting from damage to the electron transport chain complexes and mitochondrial reactive oxygen species overproduction. Conclusions This evidence clearly illustrates that microcirculatory ischemia/reperfusion injury can be attributed to Mff‐dependent mitochondrial fission via voltage‐dependent anion channel 1/hexokinase 2–mediated mitochondrial permeability transition pore opening and mitochondrial reactive oxygen species/cardiolipin involved cyt‐c release.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qinhua Jin
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Chen Shi
- Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
200
|
Khan JN, McCann GP. Cardiovascular magnetic resonance imaging assessment of outcomes in acute myocardial infarction. World J Cardiol 2017; 9:109-133. [PMID: 28289525 PMCID: PMC5329738 DOI: 10.4330/wjc.v9.i2.109] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/02/2016] [Accepted: 01/02/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular magnetic resonance (CMR) imaging uniquely characterizes myocardial and microvascular injury in acute myocardial infarction (AMI), providing powerful surrogate markers of outcomes. The last 10 years have seen an exponential increase in AMI studies utilizing CMR based endpoints. This article provides a contemporary, comprehensive review of the powerful role of CMR imaging in the assessment of outcomes in AMI. The theory, assessment techniques, chronology, importance in predicting left ventricular function and remodelling, and prognostic value of each CMR surrogate marker is described in detail. Major studies illustrating the importance of the markers are summarized, providing an up to date review of the literature base in CMR imaging in AMI.
Collapse
Affiliation(s)
- Jamal N Khan
- Jamal N Khan, Gerry P McCann, Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Cardiovascular Biomedical Research Unit, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Gerry P McCann
- Jamal N Khan, Gerry P McCann, Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Cardiovascular Biomedical Research Unit, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| |
Collapse
|