1
|
Chen T, Liu TT, Bai WL, Qi Q, Yin HS, Wang T, Jiang ZA. Impact of controlled stepwise reperfusion during primary percutaneous coronary intervention on patients with ST-elevation myocardial infarction. Clin Hemorheol Microcirc 2025; 89:217-224. [PMID: 39973434 DOI: 10.1177/13860291241304948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
ObjectiveThe aim of this study is to examine the impact of controlled stepwise reperfusion by modulating pre-dilation balloon pressure during primary percutaneous coronary interventions (PPCI) in patients with ST-elevation myocardial infarction (STEMI).MethodsConsecutive STEMI patients requiring PPCI with thrombolysis in myocardial infarction (TIMI) flow grades 0 or 1, were randomly divided into an experimental group and a control group. For the control group, the pre-dilation balloon was removed immediately after achieving antegrade perfusion beyond the lesion. The experimental group underwent stepwise reperfusion, with the balloon pressure being gradually reduced. Baseline data, intra/post-procedural PPCI data, 3-month left ventricular ejection fraction (LVEF), and major adverse cardiac events (MACE) were documented and compared between the two groups.ResultsThe control group experienced more severe symptoms during the procedure (p = 0.034), higher post-procedural corrected TIMI frame counts (p = 0.047), more significant hemodynamic changes (p = 0.031), and increased rates of ventricular tachycardia/ventricular fibrillation (p = 0.035). Additionally, they had a higher total number of arrhythmias (p = 0.017), a lower 90-min ST segment resolution rate (p = 0.045), and elevated cTNI levels one week after the procedure (p = 0.047). Three months later, the control group demonstrated a lower LVEF compared to the experimental group (p = 0.048) and a trend towards more drug-treated arrhythmias (p = 0.073). No differences were observed in other statistical results.ConclusionIn patients with STEMI undergoing PPCI, controlled stepwise reperfusion by adjusting the pre-dilation balloon pressure effectively reduces myocardial ischemia-reperfusion injury, improves myocardial perfusion, and supports the recovery of cardiac function.
Collapse
Affiliation(s)
- Tao Chen
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ting-Ting Liu
- Heart Center, Hebei General Hospital, Shijiazhuang, China
| | - Wen-Lou Bai
- Heart Center, Hebei General Hospital, Shijiazhuang, China
| | - Qi Qi
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong-Shan Yin
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Wang
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhi-An Jiang
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Babiker F, Al-Kouh A. Immunoglobulin-Mediated Cardiac Protection From Ischemia/Reperfusion Injury in Diabetic Rats Is Associated With Endothelial Nitric Oxide Synthase/Glucose Transporter-4 Signaling Pathway. J Cardiovasc Pharmacol 2024; 84:319-330. [PMID: 39240727 DOI: 10.1097/fjc.0000000000001586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/22/2024] [Indexed: 09/08/2024]
Abstract
ABSTRACT The role of intravenous immunoglobulin in protecting the diabetic heart from ischemia/reperfusion (I/R) injury is unclear. Hearts isolated from adult diabetic and nondiabetic Wistar rats (n = 8 per group) were treated with intravenous immunoglobulin (IVIG) either 2 hours before euthanasia, before ischemia, or at reperfusion. Hemodynamic data were acquired using the Isoheart software version 1.524-S. Ischemia/reperfusion (I/R) injury was evaluated by 2,3,5-triphenyltetrazolium chloride staining and troponin T levels. The levels of apoptosis markers, caspases-3/8, antioxidant enzymes, superoxide dismutase and catalase, glucose transporters, GLUT-1 and GLUT-4, phosphorylated ERK1/2, and phosphorylated eNOS were estimated by Western blotting. Proinflammatory and anti-inflammatory cytokine levels were evaluated using enzyme-linked immunosorbent assays. Intravenous immunoglobulin administration abolished the effects of I/R injury in hearts subjected to hyperglycemia when infused at reperfusion, before ischemia, or at reperfusion in 4-week diabetic rat hearts and only at reperfusion in 6-week diabetic rat hearts. IVIG infusion resulted in a significant (P < 0.05) recovery of cardiac hemodynamics and decreased infarct size. IVIG also reduced the levels of troponin T, apoptotic enzymes, and proinflammatory cytokines. IVIG significantly (P < 0.05) increased the levels of anti-inflammatory cytokines, antioxidant enzymes, GLUT-4, and phosphorylated eNOS. Intravenous immunoglobulin protected the hearts from I/R injury if infused at reperfusion in the presence of hyperglycemia, in 4- and 6-week diabetic rat hearts, and when infused before ischemia in 4-week diabetic rat hearts. IVIG exerts its cardioprotective effects associated with the upregulated phosphorylated eNOS/GLUT-4 pathway.
Collapse
Affiliation(s)
- Fawzi Babiker
- Department of Physiology, College of Medicine, Kuwait University, Safat, Kuwait
| | | |
Collapse
|
3
|
Wang J, Yang L, Wu L, Li S, Ren C, Ding Y, Wei M, Ji X, Zhao W. Direct Ischemic Postconditioning Following Stroke Thrombectomy: A Promising Therapy for Reperfusion Injury. Neurosci Bull 2024; 40:1017-1020. [PMID: 38856959 PMCID: PMC11250735 DOI: 10.1007/s12264-024-01243-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/28/2024] [Indexed: 06/11/2024] Open
Affiliation(s)
- Jing Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Lu Yang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Longfei Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Sijie Li
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Ming Wei
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300222, China.
| | - Xunming Ji
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
4
|
Cohen MV, Downey JM. Initial Despair and Current Hope of Identifying a Clinically Useful Treatment of Myocardial Reperfusion Injury: Insights Derived from Studies of Platelet P2Y 12 Antagonists and Interference with Inflammation and NLRP3 Assembly. Int J Mol Sci 2024; 25:5477. [PMID: 38791515 PMCID: PMC11122283 DOI: 10.3390/ijms25105477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Myocardial necrosis following the successful reperfusion of a coronary artery occluded by thrombus in a patient presenting with ST-elevation myocardial infarction (STEMI) continues to be a serious problem, despite the multiple attempts to attenuate the necrosis with agents that have shown promise in pre-clinical investigations. Possible reasons include confounding clinical risk factors, the delayed application of protective agents, poorly designed pre-clinical investigations, the possible effects of routinely administered agents that might unknowingly already have protected the myocardium or that might have blocked protection, and the biological differences of the myocardium in humans and experimental animals. A better understanding of the pathobiology of myocardial infarction is needed to stem this reperfusion injury. P2Y12 receptor antagonists minimize platelet aggregation and are currently part of the standard treatment to prevent thrombus formation and propagation in STEMI protocols. Serendipitously, these P2Y12 antagonists also dramatically attenuate reperfusion injury in experimental animals and are presumed to provide a similar protection in STEMI patients. However, additional protective agents are needed to further diminish reperfusion injury. It is possible to achieve additive protection if the added intervention protects by a mechanism different from that of P2Y12 antagonists. Inflammation is now recognized to be a critical factor in the complex intracellular response to ischemia and reperfusion that leads to tissue necrosis. Interference with cardiomyocyte inflammasome assembly and activation has shown great promise in attenuating reperfusion injury in pre-clinical animal models. And the blockade of the executioner protease caspase-1, indeed, supplements the protection already seen after the administration of P2Y12 antagonists. Importantly, protective interventions must be applied in the first minutes of reperfusion, if protection is to be achieved. The promise of such a combination of protective strategies provides hope that the successful attenuation of reperfusion injury is attainable.
Collapse
Affiliation(s)
- Michael V. Cohen
- The Departments of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA;
- The Departments of Medicine, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA
| | - James M. Downey
- The Departments of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, Mobile, AL 36688, USA;
| |
Collapse
|
5
|
Bergman I, Gelikas S, Wexler Y, Braver O, Boyle D, Nussinovitch U. Effect of ischaemic postconditioning on markers of myocardial injury in ST-elevation myocardial infarction: a meta-analysis. Open Heart 2024; 11:e002281. [PMID: 38286569 PMCID: PMC10826564 DOI: 10.1136/openhrt-2023-002281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/31/2023] [Indexed: 01/31/2024] Open
Abstract
OBJECTIVES This study aimed to perform a meta-analysis of the short-term impact of ischaemic postconditioning (IPoC) on myocardial injury in ST elevation myocardial infarction (STEMI) using surrogate cardiac biomarkers. METHODS Eligible studies were identified using several article databases. Randomised controlled trials published between 1 January 2000 and 1 December 2021 comparing IPoC to standard of therapy in STEMI patients were included in the search. Outcomes included surrogates of myocardial injury, specifically peak troponin, creatine-kinase (CK) and CK myoglobin binding (CK-MB) enzyme levels. RESULTS 11 articles involving 1273 patients reported on CK-MB and 8 studies involving 505 patients reported on CK. Few studies used troponin as an outcome, thus, a subanalysis of troponin dynamics was not performed. Meta-regression analysis demonstrated no significant effect of IPoC on peak CK-MB (effect size -0.41, 95% CI -1.15 to 0.34) or peak CK (effect size -0.42, 95% CI -1.20 to 0.36). Linear regression analysis demonstrated a significant correlation between a history of smoking and CK-MB in the IPoC group (p=0.038). CONCLUSIONS IPoC does not seem to protect against myocardial injury in STEMI, except possibly in smokers. These results resonate with some studies using imaging techniques to ascertain myocardial damage. More research using troponin and cardiac imaging should be pursued to better assess the effects of IPoC on cardiovascular outcomes in STEMI.
Collapse
Affiliation(s)
- Idan Bergman
- Rabin Medical Center Beilinson Hospital, Petah Tikva, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | | | - Yehuda Wexler
- Technion Israel Institute of Technology The Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Omri Braver
- Barzilai Medical Center, Ashkelon, Israel
- Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dennis Boyle
- Westchester Medical Center, Valhalla, New York, USA
| | - Udi Nussinovitch
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
6
|
Bergman I, Boyle D, Braver O, Gelikas S, Wexler Y, Omelchenko A, Assali A, Nussinovitch U. Ischemic Postconditioning Confers No Benefit to Left Ventricular Systolic Function: A Meta-Analysis of Cardiac Magnetic Resonance Imaging Results. Am J Cardiol 2023; 208:126-133. [PMID: 37837795 DOI: 10.1016/j.amjcard.2023.09.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 10/16/2023]
Abstract
Ischemic postconditioning (IPoC) is a technique suggested to reduce reperfusion injury in patients suffering acute ST-elevation myocardial infarction (STEMI), although its use is highly controversial. This meta-analysis aimed to evaluate the effect of IPoC with percutaneous coronary intervention in patients with acute STEMI, as measured by follow-up left ventricular ejection fraction (LVEF) on cardiac magnetic resonance imaging. The investigators searched PubMed, Embase, and Web of Science for all randomized controlled trials published during the last 2 decades. After the removal of duplicates, 2,021 articles from online databases had been identified using relevant search criteria. The included randomized controlled trials had studied patients with acute STEMI and Thrombolysis in Myocardial Infarction flow 0 to 1 at presentation and had measured follow-up LVEF using cardiac magnetic resonance imaging. Overall, 11 studies (n = 1,339 patients) qualified for inclusion. In each study, the control group did not differ significantly from the experimental group. The pooled data from included studies were analyzed using standardized mean difference between IPoC and control groups, and the 95% confidence interval for LVEF; the results were visualized using a forest plot. Bivariate regression analyses and 1-way analyses of LVEF coefficient ratios were done to isolate for various clinical and procedural parameters. An analysis of pooled data of the IPoC (n = 674) and control (n = 665) groups showed that IPoC did not significantly impact follow-up LVEF (using standardized mean difference 0.10, 95% confidence interval 0.00 to 0.21). Further analysis showed that IPoC did not improve follow-up LVEF when isolating for relevant clinical and procedural parameters. In conclusion, the use of IPoC as an adjunctive therapy to percutaneous coronary intervention seemingly provides no benefit to left ventricular systolic function, as quantified with cardiac magnetic resonance imaging, in patients with acute STEMI with Thrombolysis in Myocardial Infarction flow 0 to 1.
Collapse
Affiliation(s)
- Idan Bergman
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Rabin Medical Center, Petach Tikva, Israel
| | | | - Omri Braver
- Department of Cardiology, Barzilai Medical Center, Ashkelon, Israel
| | - Shaul Gelikas
- The Trauma and Combat Medicine Branch, Surgeon General's Headquarters, Israel Defense Forces, Ramat Gan, Israel
| | - Yehuda Wexler
- Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexander Omelchenko
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| | - Abid Assali
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| | - Udi Nussinovitch
- Heart Institute at the Edith Wolfson Medical Center, Holon, Israel.
| |
Collapse
|
7
|
Unzué L, García E, Del Río MR, Osende J, Teijeiro-Mestre R, Fernandez-Friera L. Local Intracoronary Fibrinolysis with Distal Occlusion: A Series of Cases Using Marinade Technique. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2023; 53S:S239-S244. [PMID: 37202333 DOI: 10.1016/j.carrev.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/11/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023]
Abstract
No-reflow phenomenon is frequent in patients with ST-segment elevation myocardial infarction (STEMI) and has proven to be a strong predictor of mortality. Local fibrinolytic infusion with distal coronary occlusion (previously described as "marinade technique") can be useful in patients with acute myocardial infarction and intraluminal thrombus refractory to aspiration enabling the local effect of the drug, directly applied inside the thrombus, while protecting the microvasculature with prolonged inflation of a distal balloon. We present the early experience of four patients with inferior acute myocardial infarction and high thrombus burden successfully treated with marinade technique in one center.
Collapse
Affiliation(s)
- Leire Unzué
- Interventional Cardiology Unit, HM CIEC, HM Hospitals, Madrid, Spain; HM CIEC, HM Hospitals, Madrid, Spain; CEU San Pablo University, Spain.
| | - Eulogio García
- Interventional Cardiology Unit, HM CIEC, HM Hospitals, Madrid, Spain; HM CIEC, HM Hospitals, Madrid, Spain
| | | | - Julio Osende
- Clinical Cardiology Unit, HM CIEC, Hospital HM Sanchinarro, HM Hospitals, Madrid, Spain; HM CIEC, HM Hospitals, Madrid, Spain; CEU San Pablo University, Spain
| | - Rodrigo Teijeiro-Mestre
- Interventional Cardiology Unit, HM CIEC, HM Hospitals, Madrid, Spain; HM CIEC, HM Hospitals, Madrid, Spain
| | - Leticia Fernandez-Friera
- HM CIEC, HM Hospitals, Madrid, Spain; CEU San Pablo University, Spain; Cardiac Imaging Unit, Spain
| |
Collapse
|
8
|
Koyama T. Postconditioning with Lactate-Enriched Blood for Reducing Lethal Reperfusion Injury in Humans. J Cardiovasc Transl Res 2023; 16:793-802. [PMID: 36939958 PMCID: PMC10480094 DOI: 10.1007/s12265-023-10372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/08/2023] [Indexed: 03/21/2023]
Abstract
Ischemic myocardium cannot survive without reperfusion. However, reperfusion of the ischemic myocardium paradoxically induces myocyte death; this phenomenon is termed lethal reperfusion injury. To date, no effective approach has been demonstrated for ST-segment elevation myocardial infarction (STEMI) in clinical settings. Recently, we demonstrated a novel approach for cardioprotection, termed postconditioning with lactate-enriched blood (PCLeB). PCLeB comprises intermittent reperfusion and timely coronary injections of lactated Ringer's solution, which is implemented at the beginning of reperfusion. This approach is aimed at reducing lethal reperfusion injury via prolonging intracellular acidosis during the early period of reperfusion, compared with the original postconditioning protocol. Patients with STEMI treated using PCLeB have reported positive outcomes. This article represents an effort, with a perspective different from current insights, toward preventing lethal reperfusion injury, in light of the historical background of reperfusion injury research. PCLeB is considered the new approach for cardioprotection.
Collapse
Affiliation(s)
- Takashi Koyama
- Department of Cardiology, Saitama Municipal Hospital, 2460 Mimuro, Midori-Ku, Saitama City, Saitama, 336-8522, Japan.
| |
Collapse
|
9
|
Birnbaum Y, Ye R, Ye Y. Aspirin Blocks the Infarct-Size Limiting Effect of Ischemic Postconditioning in the Rat. Cardiovasc Drugs Ther 2023; 37:221-224. [PMID: 34403016 DOI: 10.1007/s10557-021-07241-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Ischemic postconditioning (PostC), repetitive cycles of re-occlusion, and reperfusion of the infarct-related artery immediately after reperfusion have been shown to limit myocardial infarct size in various animal models. Yet, translating the model into the clinical setting was disappointing, several clinical trials showing neutral effect. We hypothesized that aspirin loading could explain the differences between the pre-clinical and clinical studies. METHODS Male Sprague Dawley rats were subjected to 30-min coronary artery ligation. At 25 min of ischemia, animals received intravenous aspirin (20 mg/kg) or vehicle. Upon reperfusion half of the rats were randomized to PostC (3 cycles of 10-s re-occlusion/10-s reperfusion. After 4-h reperfusion, rats were euthanized. Area at risk was assessed by blue dye and infarct size by 2,3,5-triphenyl-tetrazolium-chloride (TTC). RESULTS Body weight and the size of the ischemic area at risk were comparable among groups. Infarct size expressed as a percentage of the ischemic area at risk was significantly smaller in the PostC group (13.9 ± 0.4%; p < 0.001) compared to the control group (31.0 ± 2.2%). Aspirin alone had no effect on infarct size (29.0 ± 2.6%). Yet, aspirin completely blocked the protective effect of PostC (33.3 ± 1.1%). CONCLUSIONS Aspirin, administered before reperfusion, blocks the infarct size limiting effects of PostC in the rat.
Collapse
Affiliation(s)
- Yochai Birnbaum
- The Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Regina Ye
- University of Texas At Austin, Austin, TX, USA
| | - Yumei Ye
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, BSB 648, Galveston, TX, 77555, USA.
| |
Collapse
|
10
|
Al-Kouh A, Babiker F, Al-Bader M. Renin-Angiotensin System Antagonism Protects the Diabetic Heart from Ischemia/Reperfusion Injury in Variable Hyperglycemia Duration Settings by a Glucose Transporter Type 4-Mediated Pathway. Pharmaceuticals (Basel) 2023; 16:238. [PMID: 37259385 PMCID: PMC9967344 DOI: 10.3390/ph16020238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a risk factor for cardiovascular diseases, specifically, the ischemic heart diseases (IHD). The renin-angiotensin system (RAS) affects the heart directly and indirectly. However, its role in the protection of the heart against I/R injury is not completely understood. The aim of the current study was to evaluate the efficacy of the angiotensin-converting enzyme (ACE) inhibitor and Angiotensin II receptor (AT1R) blocker or a combination thereof in protection of the heart from I/R injury. METHODS Hearts isolated from adult male Wistar rats (n = 8) were subjected to high glucose levels; acute hyperglycemia or streptozotocin (STZ)-induced diabetes were used in this study. Hearts were subjected to I/R injury, treated with Captopril, an ACE inhibitor; Losartan, an AT1R antagonist; or a combination thereof. Hemodynamics data were measured using a suitable software for that purpose. Additionally, infarct size was evaluated using 2,3,5-Triphenyltetrazolium chloride (TTC) staining. The levels of apoptosis markers (caspase-3 and -8), antioxidant enzymes, superoxide dismutase (SOD) and catalase (CAT), nitric oxide synthase (eNOS), and glucose transporter type 4 (GLUT-4) protein levels were evaluated by Western blotting. Pro-inflammatory and anti-inflammatory cytokines levels were evaluated by enzyme-linked immunosorbent assay (ELISA). RESULTS Captopril and Losartan alone or in combination abolished the effect of I/R injury in hearts subjected to acute hyperglycemia or STZ-induced diabetes. There was a significant (p < 0.05) recovery in hemodynamics, infarct size, and apoptosis markers following the treatment with Captopril, Losartan, or their combination. Treatment with Captopril, Losartan, or their combination significantly (p < 0.05) reduced pro-inflammatory cytokines and increased GLUT-4 protein levels. CONCLUSIONS The blockade of the RAS system protected the diabetic heart from I/R injury. This protection followed a pathway that utilizes GLUT-4 to decrease the apoptosis markers, pro-inflammatory cytokines, and to increase the anti-inflammatory cytokines. This protection seems to employ a pathway which is not involving ERK1/2 and eNOS.
Collapse
Affiliation(s)
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Kuwait University, P.O. Box 24923, Kuwait City 13110, Kuwait
| | | |
Collapse
|
11
|
A Novel Reperfusion Strategy for Primary Percutaneous Coronary Intervention in Patients with Acute ST-Segment Elevation Myocardial Infarction: A Prospective Case Series. J Clin Med 2023; 12:jcm12020433. [PMID: 36675362 PMCID: PMC9864309 DOI: 10.3390/jcm12020433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Ischemia reperfusion injury (IRI) remains a major problem in patients with acute ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PCI). We have developed a novel reperfusion strategy for PCI and named it "volume-controlled reperfusion (VCR)". The aim of the current study was to assess the safety and feasibility of VCR in patients with STEMI. METHODS Consecutive patients admitted to Beijing Chaoyang Hospital with STEMI were prospectively enrolled. The feasibility endpoint was procedural success. The safety endpoints included death from all causes, major vascular complications, and major adverse cardiac event (MACE), i.e., a composite of cardiac death, myocardial reinfarction, target vessel revascularization (TVR), and heart failure. RESULTS A total of 30 patients were finally included. Procedural success was achieved in 28 (93.3%) patients. No patients died during the study and no major vascular complications or MACE occurred during hospitalization. With the exception of one patient (3.3%) who underwent TVR three months after discharge, no patient encountered death (0.0%), major vascular complications (0.0%), or and other MACEs (0.0%) during the median follow-up of 16 months. CONCLUSION The findings of the pilot study suggest that VCR has favorable feasibility and safety in patients with STEMI. Further larger randomized trials are required to evaluate the effectiveness of VCR in STEMI patients.
Collapse
|
12
|
Ferdinandy P, Andreadou I, Baxter GF, Bøtker HE, Davidson SM, Dobrev D, Gersh BJ, Heusch G, Lecour S, Ruiz-Meana M, Zuurbier CJ, Hausenloy DJ, Schulz R. Interaction of Cardiovascular Nonmodifiable Risk Factors, Comorbidities and Comedications With Ischemia/Reperfusion Injury and Cardioprotection by Pharmacological Treatments and Ischemic Conditioning. Pharmacol Rev 2023; 75:159-216. [PMID: 36753049 PMCID: PMC9832381 DOI: 10.1124/pharmrev.121.000348] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/07/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Preconditioning, postconditioning, and remote conditioning of the myocardium enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and the potential to provide novel therapeutic paradigms for cardioprotection. While many signaling pathways leading to endogenous cardioprotection have been elucidated in experimental studies over the past 30 years, no cardioprotective drug is on the market yet for that indication. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic preclinical evaluation of promising cardioprotective therapies prior to their clinical evaluation, since ischemic heart disease in humans is a complex disorder caused by or associated with cardiovascular risk factors and comorbidities. These risk factors and comorbidities induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury and responses to cardioprotective interventions. Moreover, some of the medications used to treat these comorbidities may impact on cardioprotection by again modifying cellular signaling pathways. The aim of this article is to review the recent evidence that cardiovascular risk factors as well as comorbidities and their medications may modify the response to cardioprotective interventions. We emphasize the critical need for taking into account the presence of cardiovascular risk factors as well as comorbidities and their concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple comorbidities. SIGNIFICANCE STATEMENT: Ischemic heart disease is a major cause of mortality; however, there are still no cardioprotective drugs on the market. Most studies on cardioprotection have been undertaken in animal models of ischemia/reperfusion in the absence of comorbidities; however, ischemic heart disease develops with other systemic disorders (e.g., hypertension, hyperlipidemia, diabetes, atherosclerosis). Here we focus on the preclinical and clinical evidence showing how these comorbidities and their routine medications affect ischemia/reperfusion injury and interfere with cardioprotective strategies.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Ioanna Andreadou
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Hans Erik Bøtker
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sean M Davidson
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Dobromir Dobrev
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Bernard J Gersh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sandrine Lecour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Marisol Ruiz-Meana
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Coert J Zuurbier
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|
13
|
Emakhu J, Monplaisir L, Aguwa C, Arslanturk S, Masoud S, Nassereddine H, Hamam MS, Miller JB. Acute coronary syndrome prediction in emergency care: A machine learning approach. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 225:107080. [PMID: 36037605 DOI: 10.1016/j.cmpb.2022.107080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 07/30/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Clinical concern for acute coronary syndrome (ACS) is one of emergency medicine's most common patient encounters. This study aims to develop an ensemble learning-driven framework as a diagnostic support tool to prevent misdiagnosis. METHODS We obtained extensive clinical electronic health data on patient encounters with clinical concerns for ACS from a large urban emergency department (ED) between January 2017 and August 2020. We applied an analytical framework equipped with many well-developed algorithms to improve the data quality by addressing missing values, dimensionality reduction, and data imbalance. We trained ensemble learning algorithms to classify patients with ACS or non-ACS etiologies of their symptoms. We used performance evaluation metrics such as accuracy, sensitivity, precision, F1-score, and the area under the receiver operating characteristic (AUROC) to measure the model's performance. RESULTS The analysis included 31,228 patients, of whom 563 (1.8%) had ACS and 30,665 (98.2%) had alternative diagnoses. Eleven features, including systolic blood pressure, brain natriuretic peptide, chronic heart disease, coronary artery disease, creatinine, glucose, heart attack, heart rate, nephrotic syndrome, red cell distribution width, and troponin level, are reported as significantly contributing risk factors. The proposed framework successfully classifies these cohorts with sensitivity and AUROC as high as 86.3% and 93.3%. Our proposed model's accuracy, precision, specificity, Matthew's correlation coefficient, and F1-score were 85.7%, 86.3%, 93%, 80%, and 86.3%, respectively. CONCLUSION Our proposed framework can identify early patients with ACS through further refinement and validation.
Collapse
Affiliation(s)
- Joshua Emakhu
- Department of Industrial and Systems Engineering, Wayne State University, Detroit, MI 48201, USA.
| | - Leslie Monplaisir
- Department of Industrial and Systems Engineering, Wayne State University, Detroit, MI 48201, USA
| | - Celestine Aguwa
- Department of Industrial and Systems Engineering, Wayne State University, Detroit, MI 48201, USA
| | - Suzan Arslanturk
- Department of Computer Science, Wayne State University, Detroit, MI 48202, USA
| | - Sara Masoud
- Department of Industrial and Systems Engineering, Wayne State University, Detroit, MI 48201, USA
| | | | - Mohamed S Hamam
- Emergency Department, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Joseph B Miller
- Emergency Department, Henry Ford Hospital, Detroit, MI 48202, USA
| |
Collapse
|
14
|
Gao W, Zhong X, Ma Y, Huang D, Wang R, Zhao S, Yang S, Qian J, Ge J. A randomized multicenter trial to evaluate early invasive strategy for patients with acute ST-segment elevation myocardial infarction presenting 24-48 hours from symptom onset: Protocol of the RESCUE-MI study. Am Heart J 2022; 251:54-60. [PMID: 35525262 DOI: 10.1016/j.ahj.2022.05.002] [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: 12/20/2021] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND For ST-segment elevation myocardial infarction (STEMI) patients presenting 24 to 48 hours from symptom onset, whether early invasive strategy should be performed still remains controversial. METHODS This is a prospective, open-label, multicenter, investigator initiated, randomized controlled trial (NCT04962178) to evaluate the efficacy of early invasive strategy for STEMI patients within 24 to 48 hours of symptom onset. A total of 366 patients will be included from 10 hospitals in mainland China. They will be randomly (1:1) divided into 2 groups: the early invasive strategy group (primary percutaneous coronary intervention, PPCI) and conservative strategy group (optimal medical therapy with primary PCI not performed). All patients will be followed for 1 month. The primary end point is myocardial infarction size on cardiac magnetic resonance (CMR). The secondary end points are as follows: (1) major adverse cardiovascular events (MACE), which is defined as a composite of cardiac death, recurrent myocardial infarction, ischemic driven target vessel revascularization and stroke; (2) other CMR end points, including microvascular obstruction, intramyocardial hemorrhage, myocardial area at risk, left ventricular ejection fraction, left ventricular end diastolic volume and left ventricular end systolic volume. DISCUSSION This study is designed to evaluate the efficacy of early invasive strategy for STEMI patients within 24 to 48 hours of symptom onset and will add more evidence for clinical practice. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04962178. Registered on July 14, 2021.
Collapse
Affiliation(s)
- Wei Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xin Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yuanji Ma
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Dong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ruochen Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Shihai Zhao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Shan Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
15
|
Discrepancy between the Actions of Glucagon-like Peptide-1 Receptor Ligands in the Protection of the Heart against Ischemia Reperfusion Injury. Pharmaceuticals (Basel) 2022; 15:ph15060720. [PMID: 35745639 PMCID: PMC9228343 DOI: 10.3390/ph15060720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022] Open
Abstract
Tirzepatide is a dual glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonist and a promising therapy for type 2 diabetes mellitus (T2DM). GLP-1 is an incretin hormone with therapeutic potential beyond type 2 diabetes mellitus. However, GLP-1 is rapidly degraded by dipeptdyl peptidase-IV (DPP-IV) to GLP-1 (9-36). Exendin-4 (Ex-4) is a DPP-IV-resistant GLP-1 receptor agonist which, when truncated to Ex-4 (9-39), acts as a GLP-1 receptor antagonist. In the present study, hearts isolated from Wistar rats (n = 8 per group) were perfused with a modified Langendorff preparation. Left ventricular (LV) contractility and cardiovascular hemodynamics were evaluated by a data acquisition program and infarct size was evaluated by 2,3,5-Triphenyl-2H-tetrazolium chloride (TTC) staining and cardiac enzyme levels. Hearts were subjected to 30 min regional ischemia, produced by ligation of the left anterior descending (LAD) coronary artery followed by 30 min reperfusion. Hearts were treated during reperfusion with either the non-lipidated precursor of tirzepatide (NLT), GLP-1, GLP-1 (9-36), or Ex-4 in the presence or absence of Ex-4 (9-39). Infusion of GLP-1 (9-36) or Ex-4 protected the heart against I/R injury (p > 0.01) by normalizing cardiac hemodynamic and enzyme levels. Neither GLP-1, NLT, nor Ex-4 (9-39) showed any protection. Interestingly, Ex-4 (9-39) blocked Ex-4-mediated protection but not that of GLP-1 (9-36). These data suggest that Ex-4-mediated protection is GLP-1-receptor-dependent but GLP-1 (9-36)-mediated protection is not.
Collapse
|
16
|
Madsen JM, Glinge C, Jabbari R, Nepper-Christensen L, Høfsten DE, Tilsted HH, Holmvang L, Pedersen F, Joshi FR, Sørensen R, Bang LE, Bøtker HE, Terkelsen CJ, Mæng M, Jensen LO, Aarøe J, Kelbæk H, Torp-Pedersen C, Køber L, Lønborg JT, Engstrøm T. Comparison of Effect of Ischemic Postconditioning on Cardiovascular Mortality in Patients With ST-Segment Elevation Myocardial Infarction Treated With Primary Percutaneous Coronary Intervention With Versus Without Thrombectomy. Am J Cardiol 2022; 166:18-24. [PMID: 34930614 DOI: 10.1016/j.amjcard.2021.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
In patients with ST-segment elevation myocardial infarction (STEMI), ischemic postconditioning (iPOST) have shown ambiguous results in minimizing reperfusion injury. Previous findings show beneficial effects of iPOST in patients with STEMI treated without thrombectomy. However, it remains unknown whether the cardioprotective effect of iPOST in these patients persist on long term. In the current study, all patients were identified through the DANAMI-3-iPOST database. Patients were randomized to conventional primary percutaneous coronary intervention (PCI) or iPOST in addition to PCI. Cumulative incidence rates were calculated, and multivariable analyses stratified according to thrombectomy use were performed. The primary end point was a combination of cardiovascular mortality and hospitalization for heart failure. From 2011 to 2014, 1,234 patients with STEMI were included with a median follow-up of 4.8 years. In patients treated without thrombectomy (n = 520), the primary end point occurred in 15% (48/326) in the iPOST group and in 22% (42/194) in the conventional group (unadjusted hazard ratio [HR] 0.62, 95% confidence interval [CI] 0.41 to 0.94, p = 0.023). In adjusted Cox analysis, iPOST remained associated with reduced long-term risk of cardiovascular mortality (HR 0.53, 95% CI 0.29 to 0.97, p = 0.039). In patients treated with thrombectomy (n = 714), there was no significant difference between iPOST (17%, 49/291) and conventional treatment (17%, 72/423) on the primary end point (unadjusted HR 1.01, 95% CI 0.70 to 1.45, p = 0.95). During a follow-up of nearly 5 years, iPOST reduced long-term occurrence of cardiovascular mortality and hospitalization for heart failure in patients with STEMI treated with PCI but without thrombectomy.
Collapse
|
17
|
Ye R, Jneid H, Alam M, Uretsky BF, Atar D, Kitakaze M, Davidson SM, Yellon DM, Birnbaum Y. Do We Really Need Aspirin Loading for STEMI? Cardiovasc Drugs Ther 2022; 36:1221-1238. [PMID: 35171384 DOI: 10.1007/s10557-022-07327-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
Abstract
Aspirin loading (chewable or intravenous) as soon as possible after presentation is a class I recommendation by current ST elevation myocardial infarction (STEMI) guidelines. Earlier achievement of therapeutic antiplatelet effects by aspirin loading has long been considered the standard of care. However, the effects of the loading dose of aspirin (alone or in addition to a chronic maintenance oral dose) have not been studied. A large proportion of myocardial cell death occurs upon and after reperfusion (reperfusion injury). Numerous agents and interventions have been shown to limit infarct size in animal models when administered before or immediately after reperfusion. However, these interventions have predominantly failed to show significant protection in clinical studies. In the current review, we raise the hypothesis that aspirin loading may be the culprit. Data obtained from animal models consistently show that statins, ticagrelor, opiates, and ischemic postconditioning limit myocardial infarct size. In most of these studies, aspirin was not administered. However, when aspirin was administered before reperfusion (as is the case in the majority of studies enrolling STEMI patients), the protective effects of statin, ticagrelor, morphine, and ischemic postconditioning were attenuated, which can be plausibly attributable to aspirin loading. We therefore suggest studying the effects of aspirin loading before reperfusion on the infarct size limiting effects of statins, ticagrelor, morphine, and/ or postconditioning in large animal models using long reperfusion periods (at least 24 h). If indeed aspirin attenuates the protective effects, clinical trials should be conducted comparing aspirin loading to alternative antiplatelet regimens without aspirin loading in patients with STEMI undergoing primary percutaneous coronary intervention.
Collapse
Affiliation(s)
- Regina Ye
- University of Texas at Austin, Austin, TX, USA
| | - Hani Jneid
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA
| | - Mahboob Alam
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA
| | - Barry F Uretsky
- University of Arkansas for Medical Sciences, Central Arkansas Veterans Health System, Little Rock, AR, USA
| | - Dan Atar
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Masafumi Kitakaze
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Yochai Birnbaum
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA.
| |
Collapse
|
18
|
Vidal-Calés P, Cepas-Guillén PL, Brugaletta S, Sabaté M. New Interventional Therapies beyond Stenting to Treat ST-Segment Elevation Acute Myocardial Infarction. J Cardiovasc Dev Dis 2021; 8:jcdd8090100. [PMID: 34564118 PMCID: PMC8469769 DOI: 10.3390/jcdd8090100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
Myocardial infarction remains the principal cause of death in Europe. In patients with ST-segment-elevation myocardial infarction (STEMI), a promptly revascularization with primary percutaneous intervention (PCI) has transformed prognosis in the last decades. However, despite increasing successful PCI procedures, mortality has remained unchanged in recent years. Also, due to an unsatisfactory reperfusion, some patients have significant myocardial damage and suffer left ventricular adverse remodeling with reduced function—all that resulting in the onset of heart failure with all its inherent clinical and socioeconomic burden. As a consequence of longer ischemic times, distal thrombotic embolization, ischemia-reperfusion injury and microvascular dysfunction, the resultant myocardial infarct size is the major prognostic determinant in STEMI patients. The improved understanding of all the pathophysiology underlying these events has derived to the development of several novel therapies aiming to reduce infarct size and to improve clinical outcomes in these patients. In this article, based on the mechanisms involved in myocardial infarction prognosis, we review the new interventional strategies beyond stenting that may solve the suboptimal results that STEMI patients still experience.
Collapse
Affiliation(s)
- Pablo Vidal-Calés
- Institut Clínic Cardiovascular, Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; (P.V.-C.); (P.L.C.-G.); (S.B.)
| | - Pedro L. Cepas-Guillén
- Institut Clínic Cardiovascular, Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; (P.V.-C.); (P.L.C.-G.); (S.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Salvatore Brugaletta
- Institut Clínic Cardiovascular, Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; (P.V.-C.); (P.L.C.-G.); (S.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Manel Sabaté
- Institut Clínic Cardiovascular, Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; (P.V.-C.); (P.L.C.-G.); (S.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares (CIBERCV) CB16/11/00411, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-932-275-519
| |
Collapse
|
19
|
Chen W, Deng M, Wang H, Wang Y, Zhou W, Yu T. ROS‑associated mechanism of different concentrations of pinacidil postconditioning in the rat cardiac Nrf2‑ARE signaling pathway. Mol Med Rep 2021; 23:433. [PMID: 33846798 PMCID: PMC8060791 DOI: 10.3892/mmr.2021.12072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 02/25/2021] [Indexed: 01/06/2023] Open
Abstract
Previous studies have confirmed that 50 µmol/l pinacidil postconditioning (PPC) activates the nuclear factor-E2 related factor 2 (Nrf2)-antioxidant responsive element (ARE) pathway, which protects the myocardium from ischemia-reperfusion (IR) injury; however, whether this is associated with reactive oxygen species (ROS) generation remains unclear. In the present study, a Langendorff rat model of isolated myocardial IR was established to investigate the mechanism of PPC at different concentrations, as well as the association between the rat myocardial Nrf2-ARE signaling pathway and ROS. A total of 48 rats were randomly divided into the following six groups (n=8 per group): i) Normal; ii) IR iii) 10 µmol/l PPC (P10); iv) 30 µmol/l PPC (P30); v) 50 µmol/l PPC (P50); and vi) N-(2-mercaptopropionyl)-glycine (MPG; a ROS scavenger) + 50 µmol/l pinacidil (P50 + MPG). At the end of reperfusion (T3), compared with the IR group, the P10, P30 and P50 groups exhibited improved cardiac function, such as left ventricular development pressure, heart rate, left ventricular end-diastolic pressure, +dp/dtmax, myocardial cell ultrastructure and mitochondrial Flameng score. Furthermore, the P10 and P50 groups demonstrated the weakest and most marked improvements, respectively. Additionally, in the P10, P30 and P50 groups, the residual ROS content at the end of reperfusion was highly negatively correlated with relative expression levels of Nrf2 gene and protein. Higher pinacidil concentration was associated with higher ROS generation at 5 min post-reperfusion (T2), although this was significantly lower compared with the IR group, as well as with increased expression levels of antioxidant proteins and phase II detoxification enzymes downstream of the Nrf2 and Nrf2-ARE pathways. This result was associated with a stronger ability to scavenge ROS during reperfusion, leading to lower levels of ROS at the end of reperfusion (T3) and less myocardial damage. The optimal myocardial protective effect was achieved by 50 mmol/l pinacidil. However, cardiac function of the P50 + MPG group was significantly decreased, ultrastructure of cardiomyocytes was significantly impaired and the relative expression levels of genes and proteins in the Nrf2-ARE pathway were decreased. The aforementioned results confirmed that different PPC concentrations promoted early generation of ROS and activated the Nrf2-ARE signaling pathway following reperfusion, regulated expression levels of downstream antioxidant proteins and alleviated myocardial IR injury in rats. Treatment with 50 mmol/l pinacidil resulted in the best myocardial protection.
Collapse
Affiliation(s)
- Wei Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Mengyuan Deng
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Haiying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Ying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Wenjing Zhou
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Tian Yu
- Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
20
|
Penna C, Andreadou I, Aragno M, Beauloye C, Bertrand L, Lazou A, Falcão‐Pires I, Bell R, Zuurbier CJ, Pagliaro P, Hausenloy DJ. Effect of hyperglycaemia and diabetes on acute myocardial ischaemia-reperfusion injury and cardioprotection by ischaemic conditioning protocols. Br J Pharmacol 2020; 177:5312-5335. [PMID: 31985828 PMCID: PMC7680002 DOI: 10.1111/bph.14993] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/19/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic patients are at increased risk of developing coronary artery disease and experience worse clinical outcomes following acute myocardial infarction. Novel therapeutic strategies are required to protect the myocardium against the effects of acute ischaemia-reperfusion injury (IRI). These include one or more brief cycles of non-lethal ischaemia and reperfusion prior to the ischaemic event (ischaemic preconditioning [IPC]) or at the onset of reperfusion (ischaemic postconditioning [IPost]) either to the heart or to extracardiac organs (remote ischaemic conditioning [RIC]). Studies suggest that the diabetic heart is resistant to cardioprotective strategies, although clinical evidence is lacking. We overview the available animal models of diabetes, investigating acute myocardial IRI and cardioprotection, experiments investigating the effects of hyperglycaemia on susceptibility to acute myocardial IRI, the response of the diabetic heart to cardioprotective strategies e.g. IPC, IPost and RIC. Finally we highlight the effects of anti-hyperglycaemic agents on susceptibility to acute myocardial IRI and cardioprotection. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of PharmacyNational and Kapodistrian University of AthensAthensGreece
| | - Manuela Aragno
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | | | - Luc Bertrand
- Division of CardiologyCliniques Universitaires Saint‐LucBrusselsBelgium
- Pole of Cardiovascular Research, Institut de Recherche Experimetnale et CliniqueUCLouvainBrusselsBelgium
| | - Antigone Lazou
- School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Ines Falcão‐Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de MedicinaUniversidade do PortoPortoPortugal
| | - Robert Bell
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
| | - Coert J. Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam UMCUniversity of Amsterdam, Cardiovascular SciencesAmsterdamThe Netherlands
| | - Pasquale Pagliaro
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Derek J. Hausenloy
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular and Metabolic Disorders ProgramDuke–NUS Medical SchoolSingapore
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cardiovascular Research Center, College of Medical and Health SciencesAsia UniversityTaiwan
| |
Collapse
|
21
|
Comparison of infarction size, complete ST-segment resolution incidence, mortality and re-infarction and target vessel revascularization between remote ischemic conditioning and ischemic postconditioning in ST-segment elevation myocardial infarction patients undergoing primary percutaneous coronary intervention. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2020; 16:278-286. [PMID: 33597992 PMCID: PMC7863805 DOI: 10.5114/aic.2020.99262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/06/2020] [Indexed: 11/30/2022] Open
Abstract
Introduction Due to higher morbidity and mortality, ST-segment elevation myocardial infarction (STEMI) causes many public health problems. Aim To observe effects of remote ischemic conditioning (RIC) and ischemic postconditioning (IPC) on patients diagnosed as STEMI undergoing primary percutaneous coronary intervention (pPCI). Material and methods This meta-analysis was conducted using indirect comparison by conducting a network meta-analysis (NMA). We conducted searches by utilizing PubMed and the other databases to identify randomized controlled trials (RCTs) that described IPC or RIC treated patients diagnosed with STEMI during processes of pPCI. Enzymatic infarct size and infarction size were evaluated and cardiac events were assessed during the follow-up. Results Pooled results showed that lower enzymatic infarction size was associated with the RIC group compared to the IPC group (IPC vs. RIC: standardized mean difference (SMD) = 1.126; 95% confidence interval (CI): 0.756–1.677). Compared with IPC, RIC significantly reduced infarction size, which was assessed using cardiac magnetic resonance (CMR) (SMD = 1.113; 95% CI: 0.674–1.837). We noted a potential toward greater complete ST-segment resolution in RIC patients compared with IPC patients (odds ratio (OR) = 0.821; 95% CI: 0.166–4.051). No significant difference existed in all-cause mortality (OR = 2.211; 95% CI: 0.845–5.784), Target vessel revascularization (TVR) (OR = 0.045; 95% CI: 0.001–.662) or re-infarction (OR = 1.763; 95% CI: 0.741–4.193). Conclusions This meta-analysis suggested RIC was correlated with significantly smaller infarction size compared to IPC. No significant superiority between RIC and IPC has been observed in this study on cSTR incidence, mortality and re-infarction or TVR.
Collapse
|
22
|
Tehrani BN, Basir MB, Kapur NK. Acute myocardial infarction and cardiogenic shock: Should we unload the ventricle before percutaneous coronary intervention? Prog Cardiovasc Dis 2020; 63:607-622. [PMID: 32920027 DOI: 10.1016/j.pcad.2020.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022]
Abstract
Despite early reperfusion and coordinated systems of care, cardiogenic shock (CS) remains the number one cause of morbidity and in-hospital mortality following acute myocardial infarction (AMI). CS is a complex clinical syndrome that begins with hemodynamic instability and can progress to multi-organ failure and profound hemo-metabolic compromise. To improve outcomes, a clear understanding of the treatment objectives in CS and developing time-sensitive management strategies aimed at stabilizing hemodynamics and restoring myocardial perfusion are critical. Left ventricular (LV) load has been identified as an independent predictor of heart failure and mortality following AMI. Decades of preclinical and clinical research have identified several effective LV unloading strategies. Recent initiatives from single and multi-center registries and more recently the Door to Unload (DTU)-STEMI pilot study have provided valuable insight to developing a standardized treatment approach to AMI, based on early invasive hemodynamics and tailored circulatory support to unload the LV. To follow is a review of the pathophysiology and prevalence of shock, limitations of current therapies, and the pre-clinical and translational basis for incorporating LV unloading into contemporary AMI and shock care.
Collapse
Affiliation(s)
- Behnam N Tehrani
- Inova Heart and Vascular Institute, Falls Church, VA, United States of America
| | - Mir B Basir
- Henry Ford Medical Center, Detroit, MI, United States of America
| | - Navin K Kapur
- The CardioVascular Center, Tufts Medical Center, Boston, MA, United States of America.
| |
Collapse
|
23
|
He X, Wang Q, Zhao Y, Zhang H, Wang B, Pan J, Li J, Yu H, Wang L, Dai J, Wang D. Effect of Intramyocardial Grafting Collagen Scaffold With Mesenchymal Stromal Cells in Patients With Chronic Ischemic Heart Disease: A Randomized Clinical Trial. JAMA Netw Open 2020; 3:e2016236. [PMID: 32910197 PMCID: PMC7489863 DOI: 10.1001/jamanetworkopen.2020.16236] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPORTANCE Cell therapy may be helpful for cardiac disease but has been fraught with poor cell retention and survival after transplantation. OBJECTIVE To determine whether cell-laden hydrogel treatment is safe and feasible for patients with chronic ischemic heart disease (CIHD). DESIGN, SETTING, AND PARTICIPANTS This randomized, double-blind clinical trial was conducted between March 1, 2016, and August 31, 2019, at a single hospital in Nanjing, China. Among 115 eligible patients with CIHD, 50 patients with left ventricular ejection fraction of 45% or less were selected to receive elective coronary artery bypass grafting (CABG) and additionally randomized to cell-plus-collagen treatment (collagen/cell group), cell treatment alone (cell group), or a control group. Sixty-five patients were excluded because of severe comorbidities or unwillingness to participate. Forty-four participants (88%) completed the study. The last patient completed 12 months of follow-up in August 2019. Analyses were prespecified and included all patients with available data. INTERVENTIONS During CABG, patients in the collagen/cell group were treated with human umbilical cord-derived mesenchymal stromal cell (hUC-MSC)-laden collagen hydrogel intramyocardial injection, and the cell group was treated with hUC-MSCs alone. Patients in the control group underwent CABG alone. MAIN OUTCOMES AND MEASURES The primary outcome was safety of the cell-laden collagen hydrogel assessed by the incidence of serious adverse events. The secondary end point was the efficacy of treatment, according to cardiovascular magnetic resonance imaging-based left ventricular ejection fraction and infarct size. RESULTS Fifty patients (mean [SD] age, 62.6 [8.3] years; 38 men [76%]) were enrolled, of whom 18 were randomized to the collagen/cell group, 17 to the cell group, and 15 to the control group. Patient characteristics did not differ among groups at baseline. For the primary end point, no significant differences in serious adverse events, myocardial damage markers, and renal or liver function were observed among all groups after treatment; the collagen/cell and cell groups each had 1 case of hospitalization because of heart failure, and no serious adverse events were seen in the control group. At 12 months after treatment, the mean infarct size percentage change was -3.1% (95% CI, -6.20% to -0.02%; P = .05) in the collagen/cell group, 5.19% (-1.85% to 12.22%, P = .35) in the cell group, and 8.59% (-3.06% to 20.25%, P = .21) in the control group. CONCLUSIONS AND RELEVANCE This study provides, to our knowledge, the first clinical evidence that the use of collagen hydrogel is safe and feasible for cell delivery. These findings provide a basis for larger clinical studies. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02635464.
Collapse
Affiliation(s)
- Xiaojun He
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiang Wang
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yannan Zhao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - He Zhang
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Bin Wang
- Center for Clinical Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jun Pan
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jie Li
- Department of Cardiology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hongming Yu
- Department of Radiology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Liudi Wang
- Center for Clinical Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jianwu Dai
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
24
|
Giblett JP, Bulluck H. Cardioprotection for Acute MI in Light of the CONDI2/ERIC-PPCI Trial: New Targets Needed. ACTA ACUST UNITED AC 2020; 15:e13. [PMID: 32944081 PMCID: PMC7479528 DOI: 10.15420/icr.2020.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023]
Abstract
Protection against ischaemia-reperfusion injury after revascularisation in acute myocardial infarction remains an enigma. Many targets have been identified, but after the failure of the recent Effect of Remote Ischaemic Conditioning on Clinical Outcomes in ST-elevation Myocardial Infarction Patients Undergoing Primary Percutaneous Coronary Intervention (CONDI2/ERIC-PPCI) trial to show translation to clinical benefit, there is still no pharmacological or mechanical strategy that has translated to clinical practice. This article addresses the results of the CONDI2/ERIC-PPCI trial in the context of previous studies of ischaemic conditioning, and then considers the prospects for other potential targets of cardioprotection. Finally, the authors examine the pitfalls and challenges in trial design for future investigation of cardioprotective strategies. In particular, this article highlights the need for careful endpoint and patient selection, as well as the need to pay attention to the biology of cardioprotection during the study.
Collapse
Affiliation(s)
- Joel P Giblett
- Department of Cardiology, Liverpool Heart and Chest Hospital Liverpool, UK
| | | |
Collapse
|
25
|
Ramachandra CJA, Hernandez-Resendiz S, Crespo-Avilan GE, Lin YH, Hausenloy DJ. Mitochondria in acute myocardial infarction and cardioprotection. EBioMedicine 2020; 57:102884. [PMID: 32653860 PMCID: PMC7355051 DOI: 10.1016/j.ebiom.2020.102884] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Acute myocardial infarction (AMI) and the heart failure (HF) that often follows are among the leading causes of death and disability worldwide. As such, new treatments are needed to protect the myocardium against the damaging effects of the acute ischaemia and reperfusion injury (IRI) that occurs in AMI, in order to reduce myocardial infarct (MI) size, preserve cardiac function, and improve patient outcomes. In this regard, cardiac mitochondria play a dual role as arbiters of cell survival and death following AMI. Therefore, preventing mitochondrial dysfunction induced by acute myocardial IRI is an important therapeutic strategy for cardioprotection. In this article, we review the role of mitochondria as key determinants of acute myocardial IRI, and we highlight their roles as therapeutic targets for reducing MI size and preventing HF following AMI. In addition, we discuss the challenges in translating mitoprotective strategies into the clinical setting for improving outcomes in AMI patients.
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Sauri Hernandez-Resendiz
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany
| | - Gustavo E Crespo-Avilan
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany
| | - Ying-Hsi Lin
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Department of Biochemistry, Medical Faculty, Justus Liebig-University, Giessen, Germany; Yong Loo Lin School of Medicine, National University Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, UK; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan.
| |
Collapse
|
26
|
Abstract
Despite the increasing use and success of interventional coronary reperfusion strategies, morbidity and mortality from acute myocardial infarction are still substantial. Myocardial infarct size is a major determinant of prognosis in these patients. Therefore, cardioprotective strategies aim to reduce infarct size. However, a perplexing gap exists between the many preclinical studies reporting infarct size reduction with mechanical and pharmacological interventions and the poor translation into better clinical outcomes in patients. This Review revisits the pathophysiology of myocardial ischaemia-reperfusion injury, including the role of autophagy and forms of cell death such as necrosis, apoptosis, necroptosis and pyroptosis. Other cellular compartments in addition to cardiomyocytes are addressed, notably the coronary microcirculation. Preclinical and clinical research developments in mechanical and pharmacological approaches to induce cardioprotection, and their signal transduction pathways, are discussed. Additive cardioprotective interventions are advocated. For clinical translation into treatments for patients with acute myocardial infarction, who typically are of advanced age, have comorbidities and are receiving several medications, not only infarct size reduction but also attenuation of coronary microvascular obstruction, as well as longer-term targets including infarct repair and reverse remodelling, must be considered to improve patient outcomes. Future clinical trials must focus on patients who really need adjunct cardioprotection, that is, those with severe haemodynamic alterations.
Collapse
|
27
|
Babiker F, Benter IF, Akhtar S. Nanotoxicology of Dendrimers in the Mammalian Heart: ex vivo and in vivo Administration of G6 PAMAM Nanoparticles Impairs Recovery of Cardiac Function Following Ischemia-Reperfusion Injury. Int J Nanomedicine 2020; 15:4393-4405. [PMID: 32606684 PMCID: PMC7310973 DOI: 10.2147/ijn.s255202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/20/2020] [Indexed: 01/30/2023] Open
Abstract
Aim The effects of polyamidoamine (PAMAM) dendrimers on the mammalian heart are not completely understood. In this study, we have investigated the effects of a sixth-generation cationic dendrimer (G6 PAMAM) on cardiac function in control and diabetic rat hearts following ischemia-reperfusion (I/R) injury. Methods Isolated hearts from healthy non-diabetic (Ctr) male Wistar rats were subjected to ischemia and reperfusion (I/R). LV contractility and hemodynamics data were computed digitally whereas cardiac damage following I/R injury was assessed by measuring cardiac enzymes. For ex vivo acute exposure experiments, G6 PAMAM was administered during the first 10 mins of reperfusion in Ctr animals. In chronic in vivo studies, nondiabetic rats (Ctr) received either vehicle or daily i.p. injections of G6 PAMAM (40 mg/kg) for 4 weeks. Diabetic (D) animals received either vehicle or daily i.p. injections of G6 PAMAM (10, 20 or 40 mg/kg) for 4 weeks. The impact of G6 PAMAM on pacing-postconditioning (PPC) was also studied in Ctr and D rats. Results In ex vivo studies, acute administration of G6 PAMAM to isolated Ctr hearts during reperfusion dose-dependently impaired recovery of cardiac hemodynamics and vascular dynamics parameters following I/R injury. Chronic daily i.p. injections of G6 PAMAM significantly (P<0.01) impaired recovery of cardiac function following I/R injury in nondiabetic animals but this was not generally observed in diabetic animals except for CF which was impaired by about 50%. G6 PAMAM treatment completely blocked the protective effects of PPC in the Ctr animals. Conclusion Acute ex vivo or chronic in vivo treatment with naked G6 PAMAM dendrimer can significantly compromise recovery of non-diabetic hearts from I/R injury and can further negate the beneficial effects of PPC. Our findings are therefore extremely important in the nanotoxicological evaluation of G6 PAMAM dendrimers for potential clinical applications in physiological and pathological settings.
Collapse
Affiliation(s)
- Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait City, Kuwait
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus, Republic of Cyprus
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
28
|
Li J, Sun D, Li Y. Novel Findings and Therapeutic Targets on Cardioprotection of Ischemia/ Reperfusion Injury in STEMI. Curr Pharm Des 2020; 25:3726-3739. [PMID: 31692431 DOI: 10.2174/1381612825666191105103417] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Abstract
Acute ST-segment elevation myocardial infarction (STEMI) remains a leading cause of morbidity and mortality around the world. A large number of STEMI patients after the infarction gradually develop heart failure due to the infarcted myocardium. Timely reperfusion is essential to salvage ischemic myocardium from the infarction, but the restoration of coronary blood flow in the infarct-related artery itself induces myocardial injury and cardiomyocyte death, known as ischemia/reperfusion injury (IRI). The factors contributing to IRI in STEMI are complex, and microvascular obstruction, inflammation, release of reactive oxygen species, myocardial stunning, and activation of myocardial cell death are involved. Therefore, additional cardioprotection is required to prevent the heart from IRI. Although many mechanical conditioning procedures and pharmacological agents have been identified as effective cardioprotective approaches in animal studies, their translation into the clinical practice has been relatively disappointing due to a variety of reasons. With new emerging data on cardioprotection in STEMI over the past few years, it is mandatory to reevaluate the effectiveness of "old" cardioprotective interventions and highlight the novel therapeutic targets and new treatment strategies of cardioprotection.
Collapse
Affiliation(s)
- Jianqiang Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Danghui Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| |
Collapse
|
29
|
Meng Z, Gai W, Song D. Postconditioning with Nitrates Protects Against Myocardial Reperfusion Injury: A New Use for an Old Pharmacological Agent. Med Sci Monit 2020; 26:e923129. [PMID: 32516304 PMCID: PMC7299064 DOI: 10.12659/msm.923129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Early reperfusion remains the key therapy to salvage viable myocardium and must be applied as soon as possible following an acute myocardial infarction (AMI) to attenuate the ischemic insult. However, reperfusion injury may develop following reintroduction of blood and oxygen to vulnerable myocytes, which results in more severe cell death than in the preceding ischemic episode. Ischemic postconditioning (I-PostC) provides a cardioprotective effect in combination with pharmacological agents. Although nitrates have been tested in many experimental and clinical studies of acute AMI to evaluate the cardioprotective effect, few investigations have been focused on nitrates postconditioning in patients undergoing percutaneous coronary intervention (PCI). This review presents the manifestations of myocardial reperfusion injury (RI) and potential mechanisms underlying it, and provides the mechanisms involved in the cardioprotection of I-PostC. We also present a new therapeutic approach to attenuate RI by use of an ‘old’ agent – nitrates – in AMI patients.
Collapse
Affiliation(s)
- Zhu Meng
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Weili Gai
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Dalin Song
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| |
Collapse
|
30
|
Stiermaier T, Jensen JO, Rommel KP, de Waha-Thiele S, Fuernau G, Desch S, Thiele H, Eitel I. Combined Intrahospital Remote Ischemic Perconditioning and Postconditioning Improves Clinical Outcome in ST-Elevation Myocardial Infarction. Circ Res 2020; 124:1482-1491. [PMID: 30929570 DOI: 10.1161/circresaha.118.314500] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Remote ischemic conditioning (RIC) or ischemic postconditioning (PostC) may protect the myocardium from ischemia-reperfusion injury in patients with ST-segment-elevation myocardial infarction. OBJECTIVE To determine whether combined intrahospital RIC and PostC or PostC alone in addition to primary percutaneous coronary intervention (PCI) reduce long-term clinical events after ST-segment-elevation myocardial infarction. METHODS AND RESULTS The present study is a post hoc analysis of a prospective trial which randomized 696 ST-segment-elevation myocardial infarction patients with symptoms <12 hours 1:1:1 to either combined RIC and PostC in addition to primary PCI, PostC alone in addition to primary PCI, or conventional PCI (control). Three cycles of RIC were performed by inflation of an upper arm blood pressure cuff for 5 minutes followed by deflation for 5 minutes. PostC was performed after primary PCI via 4 cycles of 30 seconds balloon occlusions followed by 30 seconds of reperfusion. Major adverse cardiac events consisting of cardiac death, reinfarction, and new congestive heart failure were assessed during long-term follow-up. Follow-up data were obtained in 97% of patients in median 3.6 years after the index event (interquartile range, 2.9-4.2 years). Major adverse cardiac events occurred in 10.2% of patients in the combined RIC and PostC group and in 16.9% in the control group (odds ratio, 0.56; 95% CI, 0.32-0.97; P=0.04). The difference was driven by a significantly reduced rate of new congestive heart failure in the RIC and PostC group (2.7% versus 7.8%; odds ratio, 0.32; 95% CI, 0.13-0.84; P=0.02). In contrast, PostC alone did not reduce major adverse cardiac events compared with controls (14.1% versus 16.9%; odds ratio, 0.81; 95% CI, 0.48-1.35; P=0.41), and the reduction of new congestive heart failure was not statistically significant (3.5% versus 7.8%; odds ratio, 0.43; 95% CI, 0.18-1.03; P=0.05). CONCLUSIONS Cardioprotection by combined intrahospital RIC and PostC in addition to primary PCI significantly reduced the rate of major adverse cardiac events and new congestive heart failure after ST-segment-elevation myocardial infarction. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT02158468.
Collapse
Affiliation(s)
- Thomas Stiermaier
- From the University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), Germany (T.S., J.-O.J., S.d.W.-T., G.F., I.E.).,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany (T.S., J.-O.J., S.d.W.-T., G.F., S.D., I.E.)
| | - Jan-Oluf Jensen
- From the University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), Germany (T.S., J.-O.J., S.d.W.-T., G.F., I.E.).,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany (T.S., J.-O.J., S.d.W.-T., G.F., S.D., I.E.)
| | - Karl-Philipp Rommel
- Heart Center Leipzig, University Hospital, Department of Internal Medicine/Cardiology, Germany (K.-P.R., S.D., H.T.)
| | - Suzanne de Waha-Thiele
- From the University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), Germany (T.S., J.-O.J., S.d.W.-T., G.F., I.E.).,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany (T.S., J.-O.J., S.d.W.-T., G.F., S.D., I.E.)
| | - Georg Fuernau
- From the University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), Germany (T.S., J.-O.J., S.d.W.-T., G.F., I.E.).,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany (T.S., J.-O.J., S.d.W.-T., G.F., S.D., I.E.)
| | - Steffen Desch
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany (T.S., J.-O.J., S.d.W.-T., G.F., S.D., I.E.).,Heart Center Leipzig, University Hospital, Department of Internal Medicine/Cardiology, Germany (K.-P.R., S.D., H.T.)
| | - Holger Thiele
- Heart Center Leipzig, University Hospital, Department of Internal Medicine/Cardiology, Germany (K.-P.R., S.D., H.T.)
| | - Ingo Eitel
- From the University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), Germany (T.S., J.-O.J., S.d.W.-T., G.F., I.E.).,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany (T.S., J.-O.J., S.d.W.-T., G.F., S.D., I.E.)
| |
Collapse
|
31
|
Barzyc A, Łysik W, Słyk J, Kuszewski M, Zarębiński M, Wojciechowska M, Cudnoch-Jędrzejewska A. Reperfusion injury as a target for diminishing infarct size. Med Hypotheses 2020; 137:109558. [PMID: 31958650 DOI: 10.1016/j.mehy.2020.109558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/15/2019] [Accepted: 01/07/2020] [Indexed: 12/30/2022]
Abstract
Therapies for preventing reperfusion injury (RI) have been widely studied. However, the attempts to transfer cardioprotective therapies for reducing RI from experiments into clinical practice have been so far unsuccessful. Pathophysiological mechanisms of RI are complicated and compose of many pathways e.g. hypercontracture-mediated sarcolemma rupture, mitochondrial permeability transition pore persistent opening, reactive oxygen species formation, inflammation and no-reflow phenomenon. Based on research, it cannot be determined which mechanism dominates, probably they cooperate with a domination of one or another in different clinical circumstances. Our hypothesis is, that only intervention that at the same time interferes with different (all?) pathways of RI may turn out to be effective in decreasing the final area of infarction.
Collapse
Affiliation(s)
- A Barzyc
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - W Łysik
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - J Słyk
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - M Kuszewski
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - M Zarębiński
- Independent Public Specialist Western Hospital John Paul II in Grodzisk Mazowiecki, Poland
| | - M Wojciechowska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Independent Public Specialist Western Hospital John Paul II in Grodzisk Mazowiecki, Poland.
| | - A Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
32
|
Paccalet A, Tessier N, Paillard M, Païta L, Gomez L, Gallo-Bona N, Chouabe C, Léon C, Badawi S, Harhous Z, Ovize M, Crola Da Silva C. An innovative sequence of hypoxia-reoxygenation on adult mouse cardiomyocytes in suspension to perform multilabeling analysis by flow cytometry. Am J Physiol Cell Physiol 2019; 318:C439-C447. [PMID: 31875695 DOI: 10.1152/ajpcell.00393.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide. Although major therapeutic progress has been made during the past decades, a better understanding of the underlying mechanisms will certainly help to improve patient's prognosis. In vitro models, particularly adult mouse cardiomyocytes, have been largely used; however, their fragility and large size are major obstacles to the use of flow cytometry. Conventional techniques, such as cell imaging, require the use of large numbers of animals and are time consuming. Here, we described a new, simple, and rapid one-day protocol using living adult mouse cardiomyocytes in suspension exposed to hypoxia-reoxygenation that allows a multilabeling analysis by flow cytometry. Several parameters can be measured by fluorescent probes labeling to assess cell viability (propidium iodide, calcein-AM, and Sytox Green), mitochondrial membrane potential [DilC1(5) and TMRM], reactive oxygen species production (MitoSOX Red), and mitochondrial mass (MitoTracker Deep Red). We address the robustness and sensitivity of our model using a cardioprotective agent, cyclosporine A. Overall, our new experimental set-up offers a high-speed quantitative multilabeling analysis of adult mouse cardiomyocytes exposed to hypoxia-reoxygenation. Our model might be interesting to investigate other cellular stresses (oxidative and inflammation) or to perform pharmacological screening.
Collapse
Affiliation(s)
- Alexandre Paccalet
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Nolwenn Tessier
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Melanie Paillard
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Lucille Païta
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Ludovic Gomez
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Noëlle Gallo-Bona
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Christophe Chouabe
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Christelle Léon
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Sally Badawi
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Zeina Harhous
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Michel Ovize
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France.,Service d'Explorations Fonctionnelles Cardiovasculaires and CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Claire Crola Da Silva
- Université Lyon, CarMeN Laboratory, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Institut National des Sciences Appliquées de Lyon, Lyon, Université Claude Bernard Lyon 1, Bron, France
| |
Collapse
|
33
|
Traverse JH, Swingen CM, Henry TD, Fox J, Wang YL, Chavez IJ, Lips DL, Lesser JR, Pedersen WR, Burke NM, Pai A, Lindberg JL, Garberich RF. NHLBI-Sponsored Randomized Trial of Postconditioning During Primary Percutaneous Coronary Intervention for ST-Elevation Myocardial Infarction. Circ Res 2019; 124:769-778. [PMID: 30602360 DOI: 10.1161/circresaha.118.314060] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RATIONALE Postconditioning at the time of primary percutaneous coronary intervention (PCI) for ST-segment-elevation myocardial infarction may reduce infarct size and improve myocardial salvage. However, clinical trials have shown inconsistent benefit. OBJECTIVE We performed the first National Heart, Lung, and Blood Institute-sponsored trial of postconditioning in the United States using strict enrollment criteria to optimize the early benefits of postconditioning and assess its long-term effects on left ventricular (LV) function. METHODS AND RESULTS We randomized 122 ST-segment-elevation myocardial infarction patients to postconditioning (4, 30 seconds PTCA [percutaneous transluminal coronary angioplasty] inflations/deflations)+PCI (n=65) versus routine PCI (n=57). All subjects had an occluded major epicardial artery (thrombolysis in myocardial infarction=0) with ischemic times between 1 and 6 hours with no evidence of preinfarction angina or collateral blood flow. Cardiac magnetic resonance imaging measured at 2 days post-PCI showed no difference between the postconditioning group and control in regards to infarct size (22.5±14.5 versus 24.0±18.5 g), myocardial salvage index (30.3±15.6% versus 31.5±23.6%), or mean LV ejection fraction. Magnetic resonance imaging at 12 months showed a significant recovery of LV ejection fraction in both groups (61.0±11.4% and 61.4±9.1%; P<0.01). Subjects randomized to postconditioning experienced more favorable remodeling over 1 year (LV end-diastolic volume =157±34 to 150±38 mL) compared with the control group (157±40 to 165±45 mL; P<0.03) and reduced microvascular obstruction ( P=0.05) on baseline magnetic resonance imaging and significantly less adverse LV remodeling compared with control subjects with microvascular obstruction ( P<0.05). No significant adverse events were associated with the postconditioning protocol and all patients but one (hemorrhagic stroke) survived through 1 year of follow-up. CONCLUSIONS We found no early benefit of postconditioning on infarct size, myocardial salvage index, and LV function compared with routine PCI. However, postconditioning was associated with improved LV remodeling at 1 year of follow-up, especially in subjects with microvascular obstruction. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT01324453.
Collapse
Affiliation(s)
- Jay H Traverse
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.).,Cardiovascular Division, The University of Minnesota School of Medicine, Minneapolis (J.H.T., C.M.S.)
| | - Cory M Swingen
- Cardiovascular Division, The University of Minnesota School of Medicine, Minneapolis (J.H.T., C.M.S.)
| | - Timothy D Henry
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Jane Fox
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Yale L Wang
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Ivan J Chavez
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Daniel L Lips
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - John R Lesser
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Wesley R Pedersen
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Nicholas M Burke
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Akila Pai
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Jana L Lindberg
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Ross F Garberich
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| |
Collapse
|
34
|
Rios-Navarro C, Marcos-Garces V, Bayes-Genis A, Husser O, Nuñez J, Bodi V. Microvascular Obstruction in ST-Segment Elevation Myocardial Infarction: Looking Back to Move Forward. Focus on CMR. J Clin Med 2019; 8:E1805. [PMID: 31661823 PMCID: PMC6912395 DOI: 10.3390/jcm8111805] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
After a myocardial infarction (MI), despite the resolution of the coronary occlusion, the deterioration of myocardial perfusion persists in a considerable number of patients. This phenomenon is known as microvascular obstruction (MVO). Initially, the focus was placed on re-establishing blood flow in the epicardial artery. Then, the observation that MVO has profound negative structural and prognostic repercussions revived interest in microcirculation. In the near future, the availability of co-adjuvant therapies (beyond timely coronary reperfusion) aimed at preventing, minimizing, and repairing MVOs and finding convincing answers to questions regarding what, when, how, and where to administer these therapies will be of utmost importance. The objective of this work is to review the state-of-the-art concepts on pathophysiology, diagnostic methods, and structural and clinical implications of MVOs in patients with ST-segment elevation MIs. Based on this knowledge we discuss previously-tested and future opportunities for the prevention and repair of MVO.
Collapse
Affiliation(s)
| | | | - Antoni Bayes-Genis
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain.
- Cardiology Department and Heart Failure Unit, Hospital Universitari Germans Trias i Pujol (Badalona) and Department of Medicine Universitat Autonoma de Barcelona, 08916 Barcelona, Spain.
| | - Oliver Husser
- Department of Cardiology, St-Johannes Hospital, 44137 Dortmund, Germany.
| | - Julio Nuñez
- Institute of Health Research INCLIVA, 46010 Valencia, Spain.
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain.
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain.
- Department of Medicine, Universidad de Valencia, 46010 Valencia, Spain.
| | - Vicente Bodi
- Institute of Health Research INCLIVA, 46010 Valencia, Spain.
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain.
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain.
- Department of Medicine, Universidad de Valencia, 46010 Valencia, Spain.
| |
Collapse
|
35
|
Yu CW. Does thrombectomy inhibit effect of ischaemic postconditioning in STEMI? True or not? Heart 2019; 106:3-5. [PMID: 31611324 PMCID: PMC6952833 DOI: 10.1136/heartjnl-2019-315389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Cheol Woong Yu
- Cardiology, Korea University Anam Hospital, Seoul, Republic of Korea
| |
Collapse
|
36
|
Nepper-Christensen L, Høfsten DE, Helqvist S, Lassen JF, Tilsted HH, Holmvang L, Pedersen F, Joshi F, Sørensen R, Bang L, Bøtker HE, Terkelsen CJ, Maeng M, Jensen LO, Aarøe J, Kelbæk H, Køber L, Engstrøm T, Lønborg J. Interaction of ischaemic postconditioning and thrombectomy in patients with ST-elevation myocardial infarction. Heart 2019; 106:24-32. [PMID: 31315939 DOI: 10.1136/heartjnl-2019-314952] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/24/2019] [Accepted: 06/21/2019] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE The Third Danish Study of Optimal Acute Treatment of Patients with ST-segment Elevation Myocardial Infarction - Ischaemic Postconditioning (DANAMI-3-iPOST) did not show improved clinical outcome in patients with ST-segment elevation myocardial infarction (STEMI) treated with ischaemic postconditioning. However, the use of thrombectomy was frequent and thrombectomy may in itself diminish the effect of ischaemic postconditioning. We evaluated the effect of ischaemic postconditioning in patients included in DANAMI-3-iPOST stratified by the use of thrombectomy. METHODS Patients with STEMI were randomised to conventional primary percutaneous coronary intervention (PCI) or ischaemic postconditioning plus primary PCI. The primary endpoint was a combination of all-cause mortality and hospitalisation for heart failure. RESULTS From March 2011 until February 2014, 1234 patients were included with a median follow-up period of 35 (interquartile range 28 to 42) months. There was a significant interaction between ischaemic postconditioning and thrombectomy on the primary endpoint (p=0.004). In patients not treated with thrombectomy (n=520), the primary endpoint occurred in 33 patients (10%) who underwent ischaemic postconditioning (n=326) and in 35 patients (18%) who underwent conventional treatment (n=194) (adjusted hazard ratio (HR) 0.55 (95%confidence interval (CI) 0.34 to 0.89), p=0.016). In patients treated with thrombectomy (n=714), there was no significant difference between patients treated with ischaemic postconditioning (n=291) and conventional PCI (n=423) on the primary endpoint (adjusted HR 1.18 (95% CI 0.62 to 2.28), p=0.62). CONCLUSIONS In this post-hoc study of DANAMI-3-iPOST, ischaemic postconditioning, in addition to primary PCI, was associated with reduced risk of all-cause mortality and hospitalisation for heart failure in patients with STEMI not treated with thrombectomy. TRIAL REGISTRATION NUMBER NCT01435408.
Collapse
Affiliation(s)
- Lars Nepper-Christensen
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Dan Eik Høfsten
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Steffen Helqvist
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jens Flensted Lassen
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Hans-Henrik Tilsted
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lene Holmvang
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Frants Pedersen
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Francis Joshi
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rikke Sørensen
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lia Bang
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | | | - Michael Maeng
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Lisette Okkels Jensen
- Department of Cardiology, Catheterisation Lab, Odense University Hospital, Odense, Denmark
| | - Jens Aarøe
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Henning Kelbæk
- Department of Cardiology, Roskilde University Hospital, Roskilde, Denmark
| | - Lars Køber
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Engstrøm
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jacob Lønborg
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
37
|
Ischemic Postconditioning (IPostC) Protects Fibrotic and Cirrhotic Rat Livers after Warm Ischemia. Can J Gastroenterol Hepatol 2019; 2019:5683479. [PMID: 31281804 PMCID: PMC6590494 DOI: 10.1155/2019/5683479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 05/14/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Decreased organ function following liver resection is a major clinical issue. The practical method of ischemic postconditioning (IPostC) has been studied in heart diseases, but no data exist regarding fibrotic livers. AIMS We aimed to determine whether IPostC could protect healthy, fibrotic, and cirrhotic livers from ischemia reperfusion injury (IRI). METHODS Fibrosis was induced in male SD rats using bile duct ligation (BDL, 4 weeks), and cirrhosis was induced using thioacetamide (TAA, 18 weeks). Fibrosis and cirrhosis were histologically confirmed using HE and EvG staining. For healthy, fibrotic, and cirrhotic livers, isolated liver perfusion with 90 min of warm ischemia was performed in three groups (each with n=8): control, IPostC 8x20 sec, and IPostC 4x60 sec. additionally, healthy livers were investigated during a follow-up study. Lactate dehydrogenase (LDH) and thromboxane B2 (TXB2) in the perfusate, as well as bile flow (healthy/TAA) and portal perfusion pressure, were measured. RESULTS LDH and TXB2 were reduced, and bile flow was increased by IPostC, mainly in total and in the late phase of reperfusion. The follow-up study showed that the perfusate derived from a postconditioned group had much less damaging potential than perfusate derived from the nonpostconditioned group. CONCLUSION IPostC following warm ischemia protects healthy, fibrotic, and cirrhotic livers against IRI. Reduced efflux of TXB2 is one possible mechanism for this effect of IPostC and increases sinusoidal microcirculation. These findings may help to improve organ function and recovery of patients after liver resection.
Collapse
|
38
|
Rossello X, Lobo-Gonzalez M, Ibanez B. Editor's Choice- Pathophysiology and therapy of myocardial ischaemia/reperfusion syndrome. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2019; 8:443-456. [PMID: 31172789 DOI: 10.1177/2048872619845283] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is a need to find interventions able to reduce the extent of injury in reperfused ST-segment elevation myocardial infarction (STEMI) beyond timely reperfusion. In this review, we summarise the clinical impact of STEMI from epidemiological, clinical and biological perspectives. We also revise the pathophysiology underlying the ischaemia/reperfusion syndrome occurring in reperfused STEMI, including the several players involved in this syndrome, such as cardiomyocytes, microcirculation and circulating cells. Interventions aimed to reduce the resultant infarct size, known as cardioprotective therapies, are extensively discussed, putting the focus on both mechanical interventions (i.e. ischaemic conditioning) and promising pharmacological therapies, such as early intravenous metoprolol, exenatide and other glucose modulators, N-acetylcysteine as well as on some other classic therapies which have failed to be translated to the clinical arena. Novel targets for evolving therapeutic interventions to ameliorate ischaemia/reperfusion injury are also discussed. Finally, we highlight the necessity to improve the study design of future randomised clinical trials in the field, as well as to select patients better who can most likely benefit from cardioprotective interventions.
Collapse
Affiliation(s)
- Xavier Rossello
- 1 Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Spain.,2 CIBER de enfermedades CardioVasculares (CIBERCV), Spain
| | - Manuel Lobo-Gonzalez
- 1 Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Spain
| | - Borja Ibanez
- 1 Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Spain.,2 CIBER de enfermedades CardioVasculares (CIBERCV), Spain.,3 Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Spain
| |
Collapse
|
39
|
Engstrøm T, Kelbæk H, Helqvist S, Høfsten DE, Kløvgaard L, Clemmensen P, Holmvang L, Jørgensen E, Pedersen F, Saunamaki K, Ravkilde J, Tilsted HH, Villadsen A, Aarøe J, Jensen SE, Raungaard B, Bøtker HE, Terkelsen CJ, Maeng M, Kaltoft A, Krusell LR, Jensen LO, Veien KT, Kofoed KF, Torp-Pedersen C, Kyhl K, Nepper-Christensen L, Treiman M, Vejlstrup N, Ahtarovski K, Lønborg J, Køber L. Effect of Ischemic Postconditioning During Primary Percutaneous Coronary Intervention for Patients With ST-Segment Elevation Myocardial Infarction: A Randomized Clinical Trial. JAMA Cardiol 2019; 2:490-497. [PMID: 28249094 DOI: 10.1001/jamacardio.2017.0022] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Ischemic postconditioning of the heart during primary percutaneous coronary intervention (PCI) induced by repetitive interruptions of blood flow to the ischemic myocardial region immediately after reopening of the infarct-related artery may limit myocardial damage. Objective To determine whether ischemic postconditioning can improve the clinical outcomes in patients with ST-segment elevation myocardial infarction (STEMI). Design, Setting, And Participants In this multicenter, randomized clinical trial, patients with onset of symptoms within 12 hours, STEMI, and thrombolysis in myocardial infarction (TIMI) grade 0-1 flow in the infarct-related artery at arrival were randomized to conventional PCI or postconditioning. Inclusion began on March 21, 2011, through February 2, 2014, and follow-up was completed on February 2, 2016. Analysis was based on intention to treat. Interventions Patients were randomly allocated 1:1 to conventional primary PCI, including stent implantation, or postconditioning performed as 4 repeated 30-second balloon occlusions followed by 30 seconds of reperfusion immediately after opening of the infarct-related artery and before stent implantation. Main Outcome and Measures A combination of all-cause death and hospitalization for heart failure. Results During the inclusion period, 1234 patients (975 men [79.0%] and 259 women [21.0%]; mean [SD] age, 62 [11] years) underwent randomization in the trial. Median follow-up was 38 months (interquartile range, 24-58 months). The primary outcome occurred in 69 patients (11.2%) who underwent conventional primary PCI and in 65 (10.5%) who underwent postconditioning (hazard ratio, 0.93; 95% CI, 0.66-1.30; P = .66). The hazard ratios were 0.75 (95% CI, 0.49-1.14; P = .18) for all-cause death and 0.99 (95% CI, 0.60-1.64; P = .96) for heart failure. Conclusions and Relevance Routine ischemic postconditioning during primary PCI failed to reduce the composite outcome of death from any cause and hospitalization for heart failure in patients with STEMI and TIMI grade 0-1 flow at arrival. Trial Registration clinicaltrials.gov Identifier: NCT01435408.
Collapse
Affiliation(s)
- Thomas Engstrøm
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henning Kelbæk
- Department of Cardiology, Roskilde Hospital, Roskilde, Denmark
| | - Steffen Helqvist
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Dan Eik Høfsten
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lene Kløvgaard
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Peter Clemmensen
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lene Holmvang
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Erik Jørgensen
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Frants Pedersen
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kari Saunamaki
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jan Ravkilde
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Hans-Henrik Tilsted
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anton Villadsen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Aarøe
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Bent Raungaard
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Hans E Bøtker
- Department of Cardiology, Skejby University Hospital, Skejby, Denmark
| | | | - Michael Maeng
- Department of Cardiology, Skejby University Hospital, Skejby, Denmark
| | - Anne Kaltoft
- Department of Cardiology, Skejby University Hospital, Skejby, Denmark
| | - Lars R Krusell
- Department of Cardiology, Skejby University Hospital, Skejby, Denmark
| | - Lisette O Jensen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Karsten T Veien
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Klaus Fuglsang Kofoed
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Kasper Kyhl
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Marek Treiman
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Vejlstrup
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kiril Ahtarovski
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Lønborg
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
40
|
Hausenloy DJ, Chilian W, Crea F, Davidson SM, Ferdinandy P, Garcia-Dorado D, van Royen N, Schulz R, Heusch G. The coronary circulation in acute myocardial ischaemia/reperfusion injury: a target for cardioprotection. Cardiovasc Res 2019; 115:1143-1155. [PMID: 30428011 PMCID: PMC6529918 DOI: 10.1093/cvr/cvy286] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/15/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
The coronary circulation is both culprit and victim of acute myocardial infarction. The rupture of an epicardial atherosclerotic plaque with superimposed thrombosis causes coronary occlusion, and this occlusion must be removed to induce reperfusion. However, ischaemia and reperfusion cause damage not only in cardiomyocytes but also in the coronary circulation, including microembolization of debris and release of soluble factors from the culprit lesion, impairment of endothelial integrity with subsequently increased permeability and oedema formation, platelet activation and leucocyte adherence, erythrocyte stasis, a shift from vasodilation to vasoconstriction, and ultimately structural damage to the capillaries with eventual no-reflow, microvascular obstruction (MVO), and intramyocardial haemorrhage (IMH). Therefore, the coronary circulation is a valid target for cardioprotection, beyond protection of the cardiomyocyte. Virtually all of the above deleterious endpoints have been demonstrated to be favourably influenced by one or the other mechanical or pharmacological cardioprotective intervention. However, no-reflow is still a serious complication of reperfused myocardial infarction and carries, independently from infarct size, an unfavourable prognosis. MVO and IMH can be diagnosed by modern imaging technologies, but still await an effective therapy. The current review provides an overview of strategies to protect the coronary circulation from acute myocardial ischaemia/reperfusion injury. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, Research & Development, London, UK
- Department of Cardiology, Barts Heart Centre, St Bartholomew’s Hospital, London, UK
| | - William Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, USA
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, F. Policlinico Gemelli—IRCCS, Università Cattolica Sacro Cuore, Roma, Italy
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - David Garcia-Dorado
- Department of Cardiology, Vascular Biology and Metabolism Area, Vall d’Hebron University Hospital and Research Institute (VHIR), Universitat Autónoma de Barcelona, Barcelona, Spain
- Instituto CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| |
Collapse
|
41
|
Effect of Ischemic Preconditioning and Postconditioning on Exosome-Rich Fraction microRNA Levels, in Relation with Electrophysiological Parameters and Ventricular Arrhythmia in Experimental Closed-Chest Reperfused Myocardial Infarction. Int J Mol Sci 2019; 20:ijms20092140. [PMID: 31052231 PMCID: PMC6540096 DOI: 10.3390/ijms20092140] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 12/11/2022] Open
Abstract
We investigated the antiarrhythmic effects of ischemic preconditioning (IPC) and postconditioning (PostC) by intracardiac electrocardiogram (ECG) and measured circulating microRNAs (miRs) that are related to cardiac conduction. Domestic pigs underwent 90-min. percutaneous occlusion of the mid left anterior coronary artery, followed by reperfusion. The animals were divided into three groups: acute myocardial infarction (AMI, n = 7), ischemic preconditioning-acute myocardial infarction (IPC-AMI) (n = 9), or AMI-PostC (n = 5). IPC was induced by three 5-min. episodes of repetitive ischemia/reperfusion cycles (rI/R) before AMI. PostC was induced by six 30-s rI/R immediately after induction of reperfusion 90 min after occlusion. Before the angiographic procedure, a NOGA endocardial mapping catheter was placed again the distal anterior ventricular endocardium to record the intracardiac electrogram (R-amplitude, ST-Elevation, ST-area under the curve (AUC), QRS width, and corrected QT time (QTc)) during the entire procedure. An arrhythmia score was calculated. Cardiac MRI was performed after one-month. IPC led to significantly lower ST-elevation, heart rate, and arrhythmia score during ischemia. PostC induced a rapid recovery of R-amplitude, decrease in QTc, and lower arrhythmia score during reperfusion. Slightly higher levels of miR-26 and miR-133 were observed in AMI compared to groups IPC-AMI and AMI-PostC. Significantly lower levels of miR-1, miR-208, and miR-328 were measured in the AMI-PostC group as compared to animals in group AMI and IPC-AMI. The arrhythmia score was not significantly associated with miRNA plasma levels. Cardiac MRI showed significantly smaller infarct size in the IPC-AMI group when compared to the AMI and AMI-PostC groups. Thus, IPC led to better left ventricular ejection fraction at one-month and it exerted antiarrhythmic effects during ischemia, whereas PostC exhibited antiarrhythmic properties after reperfusion, with significant downregulaton of ischemia-related miRNAs.
Collapse
|
42
|
Lou B, Zhou C. Letter by Lou and Zhou Regarding Article, "NHLBI-Sponsored Randomized Trial of Postconditioning During Primary Percutaneous Coronary Intervention for ST-Elevation Myocardial Infarction". Circ Res 2019; 124:e55-e56. [PMID: 30973802 DOI: 10.1161/circresaha.119.314734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Baohui Lou
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Chenghui Zhou
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Xing Z, Tang L, Huang J, Peng X, Hu X. Effects of ischaemic postconditioning on outcomes of patients with ST-segment elevation myocardial infarction who underwent primary percutaneous coronary intervention: a meta-analysis. BMJ Open 2019; 9:e022509. [PMID: 30904835 PMCID: PMC6475223 DOI: 10.1136/bmjopen-2018-022509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The aim of this meta-analysis was to evaluate the effects of ischaemic postconditioning (IPC) therapy on hard clinical endpoints in ST-segment elevation myocardial infarction (STEMI) patients who underwent primary percutaneous coronary intervention (PPCI). DESIGN Systematic review and meta-analysis to evaluate the effects of IPC on the outcomes of patients with STEMI. DATA SOURCES PubMed, Embase and the Cochrane Library were systematically searched for relevant articles published prior to May 1, 2018. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Randomised trials comparing conventional PPCI to PPCI combined with IPC in STEMI patients were included. The primary endpoint was heart failure. Secondary endpoints were all-cause mortality and major adverse cardiac events (MACE), including cardiac death, heart failure and MI. The Cochrane Reviewer's Handbook 4.2 was used to assess the risk of bias. DATA EXTRACTION AND SYNTHESIS Relevant data were extracted by two independent investigators. We derived pooled risk ratios (RRs) with random effects models. Sensitivity and subgroup analyses were performed. RESULTS Ten studies that had enrolled 3137 patients were included. PPCI combined with IPC failed to reduce heart failure (RR: 0.88, 95% CI: 0.61 to 1.26, p=0.47; absolute risk: 3.64% in the IPC group and 4.11% in the PPCI only group), all-cause mortality (RR: 0.94, 95% CI: 0.69 to 1.27, p=0.68; absolute risk: 5.07% in the IPC group and 5.27% in the PPCI onlygroup), MACE (RR: 1.05, 95% CI: 0.83 to 1.32, p=0.69; absolute risk: 9.37% in the IPC group and 8.93% in the PPCI only group), cardiac death (RR: 1.28, 95% CI: 0.85 to 1.93, p=0.24; absolute risk: 4.28% in the IPC group and 3.25% in the PPCI only group) and MI (RR: 1.08, 95% CI: 0.38 to 3.12, p=0.88; absolute risk: 3.61% in the IPC group and 3.44% in the PPCI only group). CONCLUSIONS IPC combined with PPCI does not reduce heart failure, MACE and all-cause mortality compared with traditional PPCI in patients with STEMI. TRIAL REGISTRATION NUMBER CRD42017063959.
Collapse
Affiliation(s)
- Zhenhua Xing
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Changsha, Hunan, China
| | - Liang Tang
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Changsha, Hunan, China
| | - Jiabing Huang
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Changsha, Hunan, China
| | - Xiaofan Peng
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Changsha, Hunan, China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
44
|
Lukovic D, Gugerell A, Zlabinger K, Winkler J, Pavo N, Baranyai T, Giricz Z, Varga ZV, Riesenhuber M, Spannbauer A, Traxler D, Jakab A, Garamvölgyi R, Petnehazy Ö, Pils D, Tóth L, Schulz R, Ferdinandy P, Gyöngyösi M. Transcriptional Alterations by Ischaemic Postconditioning in a Pig Infarction Model: Impact on Microvascular Protection. Int J Mol Sci 2019; 20:ijms20020344. [PMID: 30650650 PMCID: PMC6358966 DOI: 10.3390/ijms20020344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 11/16/2022] Open
Abstract
Although the application of cardioprotective ischaemia/reperfusion (I/R) stimuli after myocardial infarction (MI) is a promising concept for salvaging the myocardium, translation to a clinical scenario has not fulfilled expectations. We have previously shown that in pigs, ischaemic postconditioning (IPostC) reduces myocardial oedema and microvascular obstruction (MVO), without influencing myocardial infarct size. In the present study, we analyzed the mechanisms underlying the IPostC-induced microvascular protection by transcriptomic analysis, followed by pathway analysis. Closed-chest reperfused MI was induced by 90 min percutaneous balloon occlusion of the left anterior descending coronary artery, followed by balloon deflation in anaesthetised pigs. Animals were randomised to IPostC (n = 8), MI (non-conditioned, n = 8), or Control (sham-operated, n = 4) groups. After three hours or three days follow-up, myocardial tissue samples were harvested and subjected to RNA-seq analysis. Although the transcriptome analysis revealed similar expression between IPostC and MI in transcripts involved in cardioprotective pathways, we identified gene expression changes responding to IPostC at the three days follow-up. Focal adhesion signaling, downregulated genes participating in cardiomyopathy and activation of blood cells may have critical consequences for microvascular protection. Specific analyses of the gene subsets enriched in the endothelium of the infarcted area, revealed strong deregulation of transcriptional functional clusters, DNA processing, replication and repair, cell proliferation, and focal adhesion, suggesting sustentative function in the endothelial cell layer protection and integrity. The spatial and time-dependent transcriptome analysis of porcine myocardium supports a protective effect of IPostC on coronary microvasculature post-MI.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Alfred Gugerell
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Johannes Winkler
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Noemi Pavo
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Tamás Baranyai
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary.
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary.
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary.
| | - Martin Riesenhuber
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Andreas Spannbauer
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - András Jakab
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, A-1090 Vienna, Austria.
- Center for MR-Research, University Children's Hospital, 8032 Zurich, Switzerland.
| | - Rita Garamvölgyi
- Institute of Diagnostic Imaging and Radiation Oncology, University of Kaposvár, 7400 Kaposvár, Hungary.
| | - Örs Petnehazy
- Institute of Diagnostic Imaging and Radiation Oncology, University of Kaposvár, 7400 Kaposvár, Hungary.
| | - Dietmar Pils
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Levente Tóth
- Department of Radiology, University of Pécs, 7624 Pécs, Hungary.
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany.
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary.
- Pharmahungary Group, Graphisoft Park, 7 Záhony Street, H-1031 Budapest, Hungary.
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
45
|
Bulluck H, Chan MHH, Bryant JA, Chai P, Chawla A, Chua TS, Chung YC, Fei G, Ho HH, Ho AFW, Hoe AJ, Imran SS, Lee CH, Lim SH, Liew BW, Yun PLZ, Hock MOE, Paradies V, Roe MT, Teo L, Wong AS, Wong E, Wong PE, Watson T, Chan MY, Tan JW, Hausenloy DJ. Platelet inhibition to target reperfusion injury trial: Rationale and study design. Clin Cardiol 2018; 42:5-12. [PMID: 30421441 DOI: 10.1002/clc.23110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In ST-segment elevation myocardial infarction (STEMI) patients treated with primary percutaneous coronary intervention (PPCI), current oral P2Y12 platelet inhibitors do not provide maximal platelet inhibition at the time of reperfusion. Furthermore, administration of cangrelor prior to reperfusion has been shown in pre-clinical studies to reduce myocardial infarct (MI) size. Therefore, we hypothesize that cangrelor administered prior to reperfusion in STEMI patients will reduce the incidence of microvascular obstruction (MVO) and limit MI size in STEMI patients treated with PPCI. METHODS The platelet inhibition to target reperfusion injury (PITRI) trial, is a phase 2A, multi-center, double-blinded, randomized controlled trial, in which 210 STEMI patients will be randomized to receive either an intravenous (IV) bolus of cangrelor (30 μg/kg) followed by a 120-minute infusion (4 μg/kg/min) or matching saline placebo, initiated prior to reperfusion (NCT03102723). RESULTS The study started in October 2017 and the anticipated end date would be July 2020. The primary end-point will be MI size quantified by cardiovascular magnetic resonance (CMR) on day 3 post-PPCI. Secondary endpoints will include markers of reperfusion, incidence of MVO, MI size, and adverse left ventricular remodeling at 6 months, and major adverse cardiac and cerebrovascular events. SUMMARY The aim of the PITRI trial is to assess whether cangrelor administered prior to reperfusion would reduce acute MI size and MVO, as assessed by CMR.
Collapse
Affiliation(s)
- Heerajnarain Bulluck
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK.,Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK.,Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Mervyn H H Chan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore
| | - Jennifer A Bryant
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Ping Chai
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Ashish Chawla
- Department of Cardiology, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Terrance S Chua
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | | | - Gao Fei
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Hee H Ho
- Department of Cardiology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Andrew F W Ho
- SingHealth Emergency Medicine Residency Programme, Singapore, Singapore.,SingHealth Duke-NUS Emergency Medicine Academic Clinical Programme, Singapore, Singapore
| | - Andrew J Hoe
- Department of Cardiology, Mount Elizabeth Hospital, Novena, Singapore
| | - Syed S Imran
- Department of Cardiology, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Chi-Hang Lee
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Swee H Lim
- Department of Emergency Medicine, Singapore General Hospital, Singapore, Singapore
| | - Boon W Liew
- Department of Cardiology, Changi General Hospital, Singapore, Singapore
| | - Patrick L Z Yun
- Department of Cardiology, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Marcus O E Hock
- Department of Emergency Medicine, Singapore General Hospital, Singapore, Singapore.,Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Valeria Paradies
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Matthew T Roe
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Lynette Teo
- Department of Diagnostic Imaging, National University Hospital, Singapore, Singapore
| | - Aaron S Wong
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Evelyn Wong
- Department of Emergency Medicine, Singapore General Hospital, Singapore, Singapore
| | - Philip E Wong
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Timothy Watson
- Department of Cardiology, Mount Elizabeth Hospital, Novena, Singapore
| | - Mark Y Chan
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Jack W Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK.,Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore.,Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Centro de Biotecnologia-FEMSA, Nuevo Leon, México.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| |
Collapse
|
46
|
Shin ES, Chung JH, Hahn JY, Song YB, Kim EK, Yu CW, Bae JW, Chung WY, Choi SH, Choi JH, Bae JH, An KJ, Park JS, Oh JH, Kim SW, Hwang JY, Ryu JK, Garg S, Lim DS, Gwon HC, Park HS. The clinical impact of sex differences on ischemic postconditioning during primary percutaneous coronary intervention: a POST (the effects of postconditioning on myocardial reperfusion in patients with ST-segment elevation myocardial infarction) substudy. Heart Vessels 2018; 34:898-905. [DOI: 10.1007/s00380-018-1316-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/30/2018] [Indexed: 10/27/2022]
|
47
|
Hausenloy DJ, Botker HE, Engstrom T, Erlinge D, Heusch G, Ibanez B, Kloner RA, Ovize M, Yellon DM, Garcia-Dorado D. Targeting reperfusion injury in patients with ST-segment elevation myocardial infarction: trials and tribulations. Eur Heart J 2018; 38:935-941. [PMID: 27118196 PMCID: PMC5381598 DOI: 10.1093/eurheartj/ehw145] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Affiliation(s)
- Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore.,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital Skejby, DK-8200 Aarhus N, Denmark
| | - Thomas Engstrom
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - David Erlinge
- Department of Cardiology, Lund University, Lund, Sweden
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, USA.,Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Lyon, France.,UMR 1060 (CarMeN), Université Claude Bernard, Lyon, France
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
48
|
Zhou D, Ding J, Ya J, Pan L, Wang Y, Ji X, Meng R. Remote ischemic conditioning: a promising therapeutic intervention for multi-organ protection. Aging (Albany NY) 2018; 10:1825-1855. [PMID: 30115811 PMCID: PMC6128414 DOI: 10.18632/aging.101527] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022]
Abstract
Despite decades of formidable exploration, multi-organ ischemia-reperfusion injury (IRI) encountered, particularly amongst elderly patients with clinical scenarios, such as age-related arteriosclerotic vascular disease, heart surgery and organ transplantation, is still an unsettled conundrum that besets clinicians. Remote ischemic conditioning (RIC), delivered via transient, repetitive noninvasive IR interventions to distant organs or tissues, is regarded as an innovative approach against IRI. Based on the available evidence, RIC holds the potential of affording protection to multiple organs or tissues, which include not only the heart and brain, but also others that are likely susceptible to IRI, such as the kidney, lung, liver and skin. Neuronal and humoral signaling pathways appear to play requisite roles in the mechanisms of RIC-related beneficial effects, and these pathways also display inseparable interactions with each other. So far, several hurdles lying ahead of clinical translation that remain to be settled, such as establishment of biomarkers, modification of RIC regimen, and deep understanding of underlying minutiae through which RIC exerts its powerful function. As this approach has garnered an increasing interest, herein, we aim to encapsulate an overview of the basic concept and postulated protective mechanisms of RIC, highlight the main findings from proof-of-concept clinical studies in various clinical scenarios, and also to discuss potential obstacles that remain to be conquered. More well designed and comprehensive experimental work or clinical trials are warranted in future research to confirm whether RIC could be utilized as a non-invasive, inexpensive and efficient adjunct therapeutic intervention method for multi-organ protection.
Collapse
Affiliation(s)
- Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Equal contribution
| | - Jiayue Ding
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Equal contribution
| | - Jingyuan Ya
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Liqun Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
49
|
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
|
50
|
Pagliaro P, Femminò S, Popara J, Penna C. Mitochondria in Cardiac Postconditioning. Front Physiol 2018; 9:287. [PMID: 29632499 PMCID: PMC5879113 DOI: 10.3389/fphys.2018.00287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a pivotal role in cardioprotection. Here we report some fundamental studies which considered the role of mitochondrial components (connexin 43, mitochondrial KATP channels and mitochondrial permeability transition pore) in postconditioning cardioprotection. We briefly discuss the role of mitochondria, reactive oxygen species and gaseous molecules in postconditioning. Also the effects of anesthetics-used as cardioprotective substances-is briefly considered in the context of postconditioning. The role of mitochondrial postconditioning signaling in determining the limitation of cell death is underpinned. Issues in clinical translation are briefly considered. The aim of the present mini-review is to discuss in a historical perspective the role of main mitochondria mechanisms in cardiac postconditioning.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Jasmin Popara
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|