1
|
Tantawy M, Saad M, Hussien S, Selim G, Tamara A. Predilation in Primary Percutaneous Coronary Intervention. Interv Cardiol 2025; 20:e15. [PMID: 40342566 PMCID: PMC12060174 DOI: 10.15420/icr.2024.12] [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: 04/02/2024] [Accepted: 09/22/2024] [Indexed: 05/11/2025] Open
Abstract
Background In primary percutaneous coronary intervention (pPCI), balloon predilation is frequently carried out before stenting but there is a lack of data regarding optimal balloon size and the effect of balloon size on procedural and clinical outcomes. Aims This study compares small balloon predilation (≤50% of proximal vessel diameter) with large balloon predilation (>50% of proximal vessel diameter) in pPCI. Methods This multicentre prospective observational study included consecutive ST elevation MI (STEMI) patients undergoing pPCI at three tertiary centres in Egypt. Demographic, clinical and angiographic data were collected for all the patients. The primary outcome was the presence of no reflow at the conclusion of the procedure and secondary outcomes included procedural complications - no reflow, dissection, abrupt vessel closure, fluoroscopy time and procedural time - and clinical outcomes - in-hospital left ventricular ejection fraction (LVEF), cardiogenic shock, stent thrombosis, ventricular fibrillation, stroke, death, ST-segment resolution >50% 1 hour after PCI and LVEF at discharge. Results A total of 384 pPCI procedures were included. The small balloon group (n=222) and the large balloon group (n=162) were comparable in terms of baseline characteristics. The large balloon group had a significantly higher incidence of no reflow (n=23 [14.2%] versus n=6 [2.7%], p<0.001), procedural complications: n=31 [19.4%] versus n=10 [4.5%], p<0.001) and contrast volume (190.4 ± 40.2 ml versus 177.4 ± 29.4 ml, p=0.0003) compared to the small balloon group. ST-segment resolution >50% after PCI was more frequent in the small balloon group (n=182 [81.98%] versus n=109 [67.28%], p<0.001). Conclusion This study suggests that using a smaller balloon size for predilation in pPCI is associated with improved coronary flow, reduced procedural complications and better ST-segment resolution.
Collapse
Affiliation(s)
- Mahmoud Tantawy
- College of Medicine, Department of Cardiology, Misr University for Science and Technology Cairo, Egypt
| | - Marwan Saad
- Brown University Health Cardiovascular Institute, Division of Cardiology, Alpert Medical School Department of Medicine, Brown University Providence, RI, US
| | - Sherif Hussien
- College of Medicine, Department of Cardiology, Misr University for Science and Technology Cairo, Egypt
| | - Ghada Selim
- Department of Internal Medicine, Division of Cardiology, Ain Shams University Cairo, Egypt
| | - Ahmed Tamara
- Department of Internal Medicine, Division of Cardiology, Ain Shams University Cairo, Egypt
| |
Collapse
|
2
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
3
|
Samidurai A, Ockaili R, Cain C, Roh SK, Filippone SM, Kraskauskas D, Kukreja RC, Das A. Differential Regulation of mTOR Complexes with miR-302a Attenuates Myocardial Reperfusion Injury in Diabetes. iScience 2020; 23:101863. [PMID: 33319180 PMCID: PMC7725936 DOI: 10.1016/j.isci.2020.101863] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/07/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023] Open
Abstract
Persistent activation of mTOR (mammalian target of rapamycin) in diabetes increases the vulnerability of the heart to ischemia/reperfusion (I/R) injury. We show here that infusion of rapamycin (mTOR inhibitor) at reperfusion following ischemia reduced myocardial infarct size and apoptosis with restoration of cardiac function in type 1 diabetic rabbits. Likewise, treatment with rapamycin protected hyperglycemic human-pluripotent-stem-cells-derived cardiomyocytes (HG-hiPSC-CMs) following simulated ischemia (SI) and reoxygenation (RO). Phosphorylation of S6 (mTORC1 marker) was increased, whereas AKT phosphorylation (mTORC2 marker) and microRNA-302a were reduced with concomitant increase of its target, PTEN, following I/R injury in diabetic heart and HG-hiPSC-CMs. Rapamycin inhibited mTORC1 and PTEN, but augmented mTORC2 with restoration of miRNA-302a under diabetic conditions. Inhibition of miRNA-302a blocked mTORC2 and abolished rapamycin-induced protection against SI/RO injury in HG-hiPSC-CMs. We conclude that rapamycin attenuates reperfusion injury in diabetic heart through inhibition of PTEN and mTORC1 with restoration of miR-302a-mTORC2 signaling. miR-302a and AKT phosphorylation are suppressed in post-ischemic diabetic heart Negative regulator of insulin signaling, PTEN, is induced after ischemia reperfusion miRNA-302a-mimic abolishes ischemic injury in hyperglycemic human iPS cardiocytes Rapamycin treatment restores miR-302a-mTORC2 cardioprotective signaling in diabetes
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Ramzi Ockaili
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Chad Cain
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Sean K Roh
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Scott M Filippone
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Donatas Kraskauskas
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Box 980204, Virginia Commonwealth University Medical Center, 1101 East Marshall Street, Sanger Hall, Room 7020d & 7020b, Richmond, VA 23298-0204, USA
| |
Collapse
|
4
|
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
|
5
|
Kaur K, Singh N, Dhawan RK. Potential role of EphrinA2 receptors in postconditioning induced cardioprotection in rats. Eur J Pharmacol 2020; 883:173231. [PMID: 32589885 DOI: 10.1016/j.ejphar.2020.173231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/20/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022]
Abstract
EphA2 receptor has emerged as a novel cardioprotective target against myocardial infarction by preserving cardiac function, limiting infarct size and inflammation and enhancing cell survival via elevating phosphorylated Akt protein levels. However, the role of Eph receptors in postconditioning remains to be elucidated. Thus, the present study was designed to explore the role of EphA2 receptors in cardioprotective mechanism of postconditioning by employing Doxazosin as EphA2 receptor agonist, Lithocholic acid as antagonist and Wortmannin as specific phosphoinositide 3-kinase (PI3K) inhibitor. In Langendorff perfused isolated rat hearts, exposure of ischemia for 30 min succeeded by reperfusion for 2 h produced cardiac damage as determined by increase in size of infarct, LVDP, liberation of LDH and CK in effluent from coronary arteries. The reperfused hearts were homogenized and tissue concentrations of TBARs, reduced GSH and Catalase were determined. A marked rise in infarct size, liberation of LDH and CK in effluent and TBARs in myocardial tissue was observed in ischemic and reperfused hearts. Ischemic postconditioning comprising of 6 alternate episodes of 10 s ischemia and 10 s reperfusion and pharmacological post-conditioning by Doxazosin infusion for 5 min Before reperfusion confers significant protection against myocardial injury as manifested by remarkably decreased infarct size, levels of LDH, CK and tissue TBARs along with increase in GSH and Catalase activity. Pre-treatment of EphA2 antagonist, Lithocholic acid and PI3K inhibitor, Wortmannin attenuated the cardioprotective effect of postconditioning. Our results suggest that EphA2 receptors may be involved in postconditioning mediated cardioprotection probably through PI3K/Akt pathway.
Collapse
Affiliation(s)
- Kamaldeep Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India; Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India.
| | - Ravi K Dhawan
- Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| |
Collapse
|
6
|
Abstract
With the increasing age of the general population, medical conditions necessitating a surgical intervention will increase. Concomitant with advanced age, the prevalence of type 2 diabetes mellitus will also increase. These patients have a two- to three-fold higher risk of occurrence of cardiovascular events and are at higher risk of perioperative myocardial ischemia. This review will discuss recent advances in the field of perioperative cardioprotection and focus specifically on strategies that have aimed to protect the diabetic and the aged myocardium. This review will not deal with potential putative cardioprotective effects of opioids and anesthetic agents, as this is a very broad area that would necessitate a dedicated overview.
Collapse
Affiliation(s)
- Mona Momeni
- Department of Anesthesiology & Acute Medicine, Cliniques universitaires Saint Luc, Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle de Recherche Cardiovasculaire, Avenue Hippocrate, Brussels, 1200, Belgium
| | - Stefan De Hert
- Department of Anesthesiology & Perioperative Medicine, Ghent University Hospital, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
7
|
Ding M, Hu L, Yang H, Gao C, Zeng K, Yu M, Feng J, Qiu J, Liu C, Fu F, Li Y. Reduction of SIRT1 blunts the protective effects of ischemic post-conditioning in diabetic mice by impairing the Akt signaling pathway. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1677-1689. [PMID: 30954556 DOI: 10.1016/j.bbadis.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 01/01/2023]
Abstract
Ischemic post-conditioning (IPO) activates Akt signaling to confer cardioprotection. The responsiveness of diabetic hearts to IPO is impaired. We hypothesized that decreased cardiac SIRT1, a positive regulator of Akt, may be responsible for the impaired responsiveness of diabetic hearts to IPO-mediated cardioprotection. High-fat diet and streptozotocin-induced diabetic mice were subjected to myocardial ischemia/reperfusion (MI/R, 30 min ischemia and 180 min reperfusion) or IPO (three cycles of 10 s of reperfusion and ischemia at the onset of reperfusion). Adenoviral vectors encoding GFP or SIRT1 (Ad-SIRT1) were administered by direct injection into the left ventricular. Our results showed that IPO activated the Akt signaling pathway and reduced MI/R injury in non-diabetic hearts but not in diabetic hearts, in which reduced expression of SIRT1 and increased Akt acetylation were observed. Delivery of Ad-SIRT1 into the diabetic hearts reduced Akt acetylation and restored the cardioprotective effects of IPO by modulating Akt signaling pathway. In contrast, cardiac-specific SIRT1 knockout increased Akt acetylation and blunted the cardioprotective effects of IPO. In in vitro study, transfection with wild-type SIRT1 but not inactive mutant SIRT1 reduced the expression of Akt acetylation and restored the protective effects of hypoxic post-conditioning in high glucose-incubated cardiomyocytes. Moreover, the cardiomyocytes transfected with constitutive Akt acetylation showed repressed Akt phosphorylation and blunted protective effects against hypoxia/reoxygenation injury. These findings demonstrate that the reduction of SIRT1 blunts the protective effects of IPO by impairing Akt signaling pathway and that SIRT1 up-regulation restores IPO-mediated cardioprotection in diabetic mice via deacetylation-dependent activation of Akt signaling pathway.
Collapse
Affiliation(s)
- Mingge Ding
- Department of Cardiology, Xi'an Central Hospital, Xi'an Jiaotong University, China; Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Lang Hu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China
| | - Hongyan Yang
- School of Aerospace Medicine, Fourth Military Medical University, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China
| | - Ke Zeng
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Mingzhe Yu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Jiahao Feng
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Jihuan Qiu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China
| | - Chaoyang Liu
- School of Life Sciences, Northwest University, Xi'an 710000, China
| | - Feng Fu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China.
| | - Yan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China.
| |
Collapse
|
8
|
Circulating mediators of remote ischemic preconditioning: search for the missing link between non-lethal ischemia and cardioprotection. Oncotarget 2019; 10:216-244. [PMID: 30719216 PMCID: PMC6349428 DOI: 10.18632/oncotarget.26537] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022] Open
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of mortality and morbidity worldwide. There has been an extensive search for cardioprotective therapies to reduce myocardial ischemia-reperfusion (I/R) injury. Remote ischemic preconditioning (RIPC) is a phenomenon that relies on the body's endogenous protective modalities against I/R injury. In RIPC, non-lethal brief I/R of one organ or tissue confers protection against subsequent lethal I/R injury in an organ remote to the briefly ischemic organ or tissue. Initially it was believed to be limited to direct myocardial protection, however it soon became apparent that RIPC applied to other organs such as kidney, liver, intestine, skeletal muscle can reduce myocardial infarct size. Intriguing discoveries have been made in extending the concept of RIPC to other organs than the heart. Over the years, the underlying mechanisms of RIPC have been widely sought and discussed. The involvement of blood-borne factors as mediators of RIPC has been suggested by a number of research groups. The main purpose of this review article is to summarize the possible circulating mediators of RIPC, and recent studies to establish the clinical efficacy of these mediators in cardioprotection from lethal I/R injury.
Collapse
|
9
|
Le Corvoisier P, Gallet R, Lesault PF, Audureau E, Paul M, Ternacle J, Ghostine S, Champagne S, Arrouasse R, Bitari D, Mouillet G, Dubois-Randé JL, Berdeaux A, Ghaleh B, Deux JF, Teiger E. Intra-coronary morphine versus placebo in the treatment of acute ST-segment elevation myocardial infarction: the MIAMI randomized controlled trial. BMC Cardiovasc Disord 2018; 18:193. [PMID: 30340532 PMCID: PMC6194573 DOI: 10.1186/s12872-018-0936-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/09/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Experimental studies suggest that morphine may protect the myocardium against ischemia-reperfusion injury by activating salvage kinase pathways. The objective of this two-center, randomized, double-blind, controlled trial was to assess potential cardioprotective effects of intra-coronary morphine in patients with ST-segment elevation myocardial infarction (STEMI) referred for primary percutaneous intervention. METHODS Ninety-one patients with STEMI were randomly assigned to intracoronary morphine (1 mg) or placebo at reperfusion of the culprit coronary artery. The primary endpoint was infarct size/left ventricular mass ratio assessed by magnetic resonance imaging on day 3-5. Secondary endpoints included the areas under the curve (AUC) for troponin T and creatine kinase over three days, left ventricular ejection fraction assessed by echocardiography on days 1 and 6, and clinical outcomes. RESULTS Infarct size/left ventricular mass ratio was not significantly reduced by intracoronary morphine compared to placebo (27.2% ± 15.0% vs. 30.5% ± 10.6%, respectively, p = 0.28). Troponin T and creatine kinase AUCs were similar in the two groups. Morphine did not improve left ventricular ejection fraction on day 1 (49.7 ± 10.3% vs. 49.3 ± 9.3% with placebo, p = 0.84) or day 6 (48.5 ± 10.2% vs. 49.0 ± 8.5% with placebo, p = 0.86). The number of major adverse cardiac events, including stent thrombosis, during the one-year follow-up was similar in the two groups. CONCLUSIONS Intracoronary morphine at reperfusion did not significantly reduce infarct size or improve left ventricular systolic function in patients with STEMI. Presence of comorbidities in some patients may contribute to explain these results. TRIAL REGISTRATION ClinicalTrials.gov, NCT01186445 (date of registration: August 23, 2010).
Collapse
Affiliation(s)
- Philippe Le Corvoisier
- Department VERDI, Inserm, CIC1430, AP-HP, Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, F-94010, Creteil, France. .,Inserm, U955 team 3, F-94010, Creteil, France.
| | - Romain Gallet
- Inserm, U955 team 3, F-94010, Creteil, France.,Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | | | - Etienne Audureau
- Department of Public Health and CEPIA EA7376, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Muriel Paul
- Department of Pharmacy, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Julien Ternacle
- Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Saïd Ghostine
- Department of Cardiology, Marie-Lannelongue Hospital, F-92350, Le Plessis-Robinson, France
| | - Stéphane Champagne
- Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Raphaele Arrouasse
- Department VERDI, Inserm, CIC1430, AP-HP, Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, F-94010, Creteil, France
| | - Dalila Bitari
- Department VERDI, Inserm, CIC1430, AP-HP, Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, F-94010, Creteil, France
| | - Gauthier Mouillet
- Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Jean-Luc Dubois-Randé
- Inserm, U955 team 3, F-94010, Creteil, France.,Department of Cardiology, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | | | | | - Jean-François Deux
- Department of Radiology, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Emmanuel Teiger
- Inserm, U955 team 3, F-94010, Creteil, France.,Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| |
Collapse
|
10
|
Sutter EN, Mattlage AE, Bland MD, Cherry-Allen KM, Harrison E, Surkar SM, Gidday JM, Chen L, Hershey T, Lee JM, Lang CE. Remote Limb Ischemic Conditioning and Motor Learning: Evaluation of Factors Influencing Response in Older Adults. Transl Stroke Res 2018; 10:362-371. [PMID: 30088217 DOI: 10.1007/s12975-018-0653-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Remote limb ischemic conditioning (RLIC) is a clinically feasible method of promoting tissue protection against subsequent ischemic insult. Recent findings from our lab demonstrated that RLIC robustly enhances motor learning in young, healthy humans. The next step is to determine which individuals would receive maximum benefit from RLIC before applying these findings to clinical rehabilitation populations such as stroke. Numerous factors, such as age, sex, body mass index (BMI), and cardiovascular comorbidities may influence the response. Sixty-nine participants aged 40-80 were randomized to receive either RLIC (n = 33) or sham (n = 36) conditioning. Participants underwent seven consecutive sessions consisting of RLIC or sham conditioning with a blood pressure cuff on the upper extremity and motor training on a stability platform balance task, with two follow-up sessions. Balance change (post-test-pre-test) was compared across participants, groups, and the factors of age, sex, BMI, and comorbidities. Participants in both groups improved their performance on the balance task from pre- to post-test. Overall balance change was independently associated with age and BMI. There was no difference in balance change between RLIC and Sham groups. However, RLIC significantly enhanced balance performance in participants with no comorbidities. Compared with our previous study in young adults, middle-aged and older adults demonstrated smaller improvements on the balance task. RLIC enhanced learning in middle-aged and older adults only in the absence of pre-defined comorbidities. RLIC may be a promising tool for enhancing motor recovery, but the accumulation of comorbidity with age may decrease its effectiveness.
Collapse
Affiliation(s)
- Ellen N Sutter
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Anna E Mattlage
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Marghuretta D Bland
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Kendra M Cherry-Allen
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Elinor Harrison
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Swati M Surkar
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Jeffrey M Gidday
- Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Ling Chen
- Division of Biostatistics, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Tamara Hershey
- Department of Psychiatry, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA
| | - Catherine E Lang
- Program in Physical Therapy, Washington University School of Medicine, Campus Box 8502, 4444 Forest Park, Saint Louis, MO, 63108, USA.
| |
Collapse
|
11
|
Liu H, Fu L, Sun X, Peng W, Chen Z, Li Y. Remote ischemic conditioning improves myocardial parameters and clinical outcomes during primary percutaneous coronary intervention: a meta-analysis of randomized controlled trials. Oncotarget 2018; 9:8653-8664. [PMID: 29492224 PMCID: PMC5823569 DOI: 10.18632/oncotarget.23818] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/04/2017] [Indexed: 01/10/2023] Open
Abstract
We conducted a systematic review and meta-analysis to evaluate the effects of remote ischemic conditioning on myocardial parameters and clinical outcomes in ST segment elevation acute myocardial infarction (STEMI) patients undergoing primary percutaneous coronary intervention. Ten eligible randomized controlled trials with 1006 STEMI patients were identified. Compared with controls, remote ischemic conditioning reduced the myocardial enzyme levels (standardized mean difference =-0.86; 95% CI: -1.44 to -0.28; P = 0.004; I2 = 94.5%), and increased the incidence of complete ST-segment resolution [odds ratio (OR) = 1.74; 95% CI: 1.09 to 2.77; P = 0.02; I2 = 47.9%]. Remote ischemic conditioning patients had a lower risk of all-cause mortality (OR = 0.27; 95% CI: 0.12 to 0.62; P = 0.002; I2 = 0.0%) and lower major adverse cardiovascular and cerebrovascular events rate (OR=0.45; 95% CI: 0.27 to 0.75; P = 0.002; I2 = 0.0%). Meta-analysis suggested that remote ischemic conditioning conferred cardioprotection by reducing myocardial enzymes and increasing the incidence of complete ST-segment resolution in patients after STEMI. As a result, clinical outcomes were improved in terms of mortality and incidence of major adverse cardiovascular and cerebrovascular events.
Collapse
Affiliation(s)
- Hai Liu
- Third Department of Cardiac Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Li Fu
- Institute of Clinical Medicine, Department of Endocrinology, The Central Hospital of Loudi Affiliated to the University of South China, Loudi 417000, China
| | - Xiangke Sun
- Department of Cardiology, The Central Hospital of Loudi Affiliated to the University of South China, Loudi 417000, China
| | - Wei Peng
- Department of Cardiology, The Central Hospital of Loudi Affiliated to the University of South China, Loudi 417000, China
| | - Zhiwei Chen
- Department of Cardiology, The Central Hospital of Loudi Affiliated to the University of South China, Loudi 417000, China
| | - Yiliang Li
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Postdoctoral Research Workstation of Neurology, Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
12
|
Morano M, Angotti C, Tullio F, Gambarotta G, Penna C, Pagliaro P, Geuna S. Myocardial ischemia/reperfusion upregulates the transcription of the Neuregulin1 receptor ErbB3, but only postconditioning preserves protein translation: Role in oxidative stress. Int J Cardiol 2017; 233:73-79. [PMID: 28162790 DOI: 10.1016/j.ijcard.2017.01.122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 12/28/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Neuregulin1 (Nrg1) and its receptors ErbB are crucial for heart development and for adult heart structural maintenance and function and Nrg1 has been proposed for heart failure treatment. Infarct size is the major determinant of heart failure and the mechanism of action and the role of each ErbB receptor remain obscure, especially in the post-ischemic myocardium. We hypothesized that Nrg1 and ErbB are affected at transcriptional level early after ischemia/reperfusion (I/R) injury, and that the protective postconditioning procedure (PostC, brief cycles of ischemia/reperfusion carried out after a sustained ischemia) can influence this pathway. METHODS AND RESULTS The Langendorff's heart was used as an ex-vivo model to mimic an I/R injury in the whole rat heart; after 30min of ischemia and 2h of reperfusion, with or without PostC, Nrg1 and ErbB expression were analysed by quantitative real-time PCR and Western blot. While no changes occur for ErbB2, ErbB4 and Nrg1, an increase of ErbB3 expression occurs after I/R injury, with and without PostC. However, I/R reduces ErbB3 protein, whereas PostC preserves it. An in vitro analysis with H9c2 cells exposed to redox-stress indicated that the transient over-expression of ErbB3 alone is able to increase cell survival (MTT assay), limiting mitochondrial dysfunction (JC-1 probe) and apoptotic signals (Bax/Bcl-2 ratio). CONCLUSIONS This study suggests ErbB3 as a protective factor against death pathways activated by redox stress and supports an involvement of this receptor in the pro-survival responses.
Collapse
Affiliation(s)
- Michela Morano
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Carmelina Angotti
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy.
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Torino, Italy
| |
Collapse
|
13
|
Ho PWL, Pang WF, Szeto CC. Remote ischaemic pre-conditioning for the prevention of acute kidney injury. Nephrology (Carlton) 2017; 21:274-85. [PMID: 26370466 DOI: 10.1111/nep.12614] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/23/2015] [Accepted: 08/24/2015] [Indexed: 01/02/2023]
Abstract
Acute kidney injury (AKI) is a common complication associated with high morbidity and mortality in hospitalized patients. One potential mechanism underlying renal injury is ischaemia/reperfusion injury (IRI), which attributed the organ damage to the inflammatory and oxidative stress responses induced by a period of renal ischaemia and subsequent reperfusion. Therapeutic strategies that aim at minimizing the effect of IRI on the kidneys may prevent AKI and improve clinical outcomes significantly. In this review, we examine the technique of remote ischaemic preconditioning (rIPC), which has been shown by several trials to confer organ protection by applying transient, brief episodes of ischaemia at a distant site before a larger ischaemic insult. We provide an overview of the current clinical evidence regarding the renoprotective effect of rIPC in the key clinical settings of cardiac or vascular surgery, contrast-induced AKI, pre-existing chronic kidney disease (CKD) and renal transplantation, and discuss key areas for future research.
Collapse
Affiliation(s)
- Phoebe Wing-Lam Ho
- Carol & Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wing-Fai Pang
- Carol & Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Cheuk-Chun Szeto
- Carol & Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
14
|
Understanding pacing postconditioning-mediated cardiac protection: a role of oxidative stress and a synergistic effect of adenosine. J Physiol Biochem 2016; 73:175-185. [PMID: 27864790 DOI: 10.1007/s13105-016-0535-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022]
Abstract
We and others have demonstrated a protective role for pacing postconditioning (PPC) against ischemia/reperfusion (I/R) injury in the heart; however, the underlying mechanisms behind these protective effects are not completely understood. In this study, we wanted to further characterize PPC-mediated cardiac protection, specifically identify optimal pacing sites; examine the role of oxidative stress; and test the existence of a potential synergistic effect between PPC and adenosine. Isolated rat hearts were subjected to coronary occlusion followed by reperfusion. PPC involved three, 30 s, episodes of alternating left ventricular (LV) and right atrial (RA) pacing. Multiple pacing protocols with different pacing electrode locations were used. To test the involvement of oxidative stress, target-specific agonists or antagonists were infused at the beginning of reperfusion. Hemodynamic data were digitally recorded, and cardiac enzymes, oxidant, and antioxidant status were chemically measured. Pacing at the LV or RV but not at the heart apex or base significantly (P < 0.001) protected against ischemia-reperfusion injury. PPC-mediated protection was completely abrogated in the presence of reactive oxygen species (ROS) scavenger, ebselen; peroxynitrite (ONOO-) scavenger, uric acid; and nitric oxide synthase inhibitor, L-NAME. Nitric oxide (NO) donor, snap, however significantly (P < 0.05) protected the heart against I/R injury in the absence of PPC. The protective effects of PPC were significantly improved by adenosine. PPC-stimulated protection can be achieved by alternating LV and RA pacing applied at the beginning of reperfusion. NO, ROS, and the product of their interaction ONOO- play a significant role in PPC-induced cardiac protection. Finally, the protective effects of PPC can be synergized with adenosine.
Collapse
|
15
|
Babiker F, Al-Jarallah A, Joseph S. The Interplay between the Renin Angiotensin System and Pacing Postconditioning Induced Cardiac Protection. PLoS One 2016; 11:e0165777. [PMID: 27814397 PMCID: PMC5096684 DOI: 10.1371/journal.pone.0165777] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 10/18/2016] [Indexed: 01/20/2023] Open
Abstract
Background Accumulating evidence suggests a cardioprotective role of pacing postconditioning (PPC) maneuvers in animal models and more recently in humans. The procedure however remains to be optimized and its interaction with physiological systems remains to be further explored. The renin angiotensin system (RAS) plays a dual role in ischemia/reperfusion (I/R) injury. The interaction between RAS and PPC induced cardiac protection is however not clearly understood. We have recently demonstrated that angiotensin (1–7) via Mas receptor played a significant role in PPC mediated cardiac protection against I/R injury. Objective The objective of this study was to investigate the role of angiotensin converting enzyme (ACE)—chymase—angiotensin II (Ang II)—angiotensin receptor 1 (AT1) axes of RAS in PPC mediated cardiac protection. Methods Isolated rat hearts were subjected to I/R (control) or PPC in the presence or absence of Ang II, chymostatin (inhibitor of locally produced Ang II), ACE blocker (captopril) or AT1 antagonist (irbesartan). Hemodynamics data was computed digitally and infarct size was determined histologically using TTC staining and biochemically by measuring creatine kinase (CK) and lactate dehydrogenase levels. Results Cardiac hemodynamics were significantly (P<0.001) improved and infarct size and cardiac enzymes were significantly (P<0.001) reduced in hearts subjected to PPC relative to hearts subjected to I/R injury. Exogenous administration of Ang II did not affect I/R injury or PPC mediated protection. Nonetheless inhibition of endogenously synthesized Ang II protected against I/R induced cardiac damage yet did not block or augment the protective effects of PPC. The administration of AT1 antagonist did not alleviate I/R induced damage. Interestingly it abrogated PPC induced cardiac protection in isolated rat hearts. Finally, PPC induced protection and blockade of locally produced Ang II involved enhanced activation of ERK1/2 and Akt components of the reperfusion injury salvage kinase (RISK) pathway. Conclusions This study demonstrate a novel role of endogenously produced Ang II in mediating I/R injury and highlights the significance of AT1 signaling in PPC mediated cardiac protection in isolated rodents hearts ex vivo. The interaction between Ang II-AT1 and PPC appears to involve alterations in the activation state of ERK1/2 and Akt components of the RISK pathway.
Collapse
Affiliation(s)
- Fawzi Babiker
- Departments of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Jabriya, Kuwait
- * E-mail:
| | - Aishah Al-Jarallah
- Department of Biochemistry, Faculty of Medicine, Health Science Center, Kuwait University, Jabriya, Kuwait
| | - Shaji Joseph
- Departments of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
16
|
Discrepancy in calcium release from the sarcoplasmic reticulum and intracellular acidic stores for the protection of the heart against ischemia/reperfusion injury. J Physiol Biochem 2016; 72:495-508. [PMID: 27325083 DOI: 10.1007/s13105-016-0498-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/08/2016] [Indexed: 12/21/2022]
Abstract
We and others have demonstrated a protective effect of pacing postconditioning (PPC) against ischemia/reperfusion (I/R) injury. However, the mechanisms underlying this protection are not completely clear. In the present study, we evaluated the effects of calcium release from the sarcoplasmic reticulum (SR) and the novel intracellular acidic stores (AS). Isolated rat hearts (n = 6 per group) were subjected to coronary occlusion followed by reperfusion using a modified Langendorff system. Cardiac hemodynamics and contractility were assessed using a data acquisition program, and cardiac injury was evaluated by creatine kinase (CK) and lactate dehydrogenase (LDH) levels. Hearts were subjected to 30 min of regional ischemia, produced by ligation of the left anterior descending (LAD) coronary artery, followed by 30 min of reperfusion. The hearts were also subjected to PPC (3 cycles of 30 s of left ventricle (LV) pacing alternated with 30 s of right atrium (RA) pacing) and/or were treated during reperfusion with agonists or antagonists of release of calcium from SR or AS. PPC significantly (P < 0.05) normalized LV, contractility, and coronary vascular dynamics and significantly (P < 0.001) decreased heart enzyme levels compared to the control treatments. The blockade of SR calcium release resulted in a significant (P < 0.01) recovery in LV function and contractility and a significant reduction in CK and LDH levels (P < 0.01) when applied alone or in combination with PPC. Interestingly, the release of calcium from AS alone or in combination with PPC significantly improved LV function and contractility (P < 0.05) and significantly decreased the CK and LDH levels (P < 0.01) compared to the control treatments. An additive effect was produced when agonism of calcium release from AS or blockade of calcium release from the SR was combined with PPC. Calcium release from AS and blockade of calcium release from the SR protect the heart against I/R. Combining calcium release from acidic stores or blockade of calcium release from the SR with PPC produced a synergistic protective effect.
Collapse
|
17
|
Lejay A, Fang F, John R, Van JA, Barr M, Thaveau F, Chakfe N, Geny B, Scholey JW. Ischemia reperfusion injury, ischemic conditioning and diabetes mellitus. J Mol Cell Cardiol 2016; 91:11-22. [DOI: 10.1016/j.yjmcc.2015.12.020] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/15/2015] [Accepted: 12/20/2015] [Indexed: 01/08/2023]
|
18
|
Yang F, Xi L. Postconditioning of ischemic heart by intermittent ventricular pacing at the beginning of reperfusion: novel mechanisms and potential utilities in interventional cardiology settings. Am J Physiol Heart Circ Physiol 2015; 310:H1-3. [PMID: 26566731 DOI: 10.1152/ajpheart.00835.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Feiyan Yang
- Department of Cardiology, Central Hospital of Wuhan, Wuhan, China
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia; and
| |
Collapse
|
19
|
Yetgin T, van Kranenburg M, Ten Cate T, Duncker DJ, de Boer MJ, Diletti R, van Geuns RJM, Zijlstra F, Manintveld OC. Ischemic Postconditioning After Routine Thrombus Aspiration During Primary Percutaneous Coronary Intervention: Rationale and Design of the POstconditioning Rotterdam Trial. Catheter Cardiovasc Interv 2015; 88:508-514. [PMID: 27022882 DOI: 10.1002/ccd.26239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/31/2015] [Accepted: 08/24/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Whether ischemic postconditioning (IPOC) immediately after routine thrombus aspiration (TA) reduces infarct size (IS) in patients with ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PPCI) has not been established. STUDY DESIGN The POstconditioning Rotterdam Trial (PORT) is a dual-center, prospective, open-label, randomized trial with blinded endpoint evaluation enrolling 72 subjects with first-time STEMI, and an occluded infarct-related artery (IRA) without collaterals undergoing PPCI. Subjects are randomized 1:1 to a strategy of IPOC immediately after TA followed by stenting of the IRA or to conventional percutaneous coronary intervention (PCI), including TA followed by stenting of the IRA (controls). Cardiac magnetic resonance imaging (MRI) is performed at 3-5 days after STEMI and at 3 months. The primary endpoint is IS at 3 months measured by delayed enhancement MRI. Other secondary endpoints include MRI-derived microvascular obstruction (MVO), left ventricular ejection fraction, myocardial salvage index, enzymatic IS, ST-segment resolution, myocardial blush grade, microcirculatory resistance, inflammation markers, and clinical events through 3-month follow-up. CONCLUSIONS PORT is testing the hypothesis that adding IPOC (against lethal reperfusion injury) to TA (against distal embolization and MVO) is cardioprotective and reduces ultimate IS in STEMI patients undergoing PPCI (Dutch Trial Register identifier: NTR4040). © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tuncay Yetgin
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, ICIN-KNAW, Utrecht, the Netherlands
| | | | - Tim Ten Cate
- Department of Cardiology, UMC St. Radboud, Nijmegen, the Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, ICIN-KNAW, Utrecht, the Netherlands
| | - Menko-Jan de Boer
- Department of Cardiology, UMC St. Radboud, Nijmegen, the Netherlands
| | - Roberto Diletti
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Robert-Jan M van Geuns
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Department of Radiology, Erasmus MC, Rotterdam, the Netherlands
| | - Felix Zijlstra
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, ICIN-KNAW, Utrecht, the Netherlands
| | | |
Collapse
|
20
|
McCafferty K, Forbes S, Thiemermann C, Yaqoob MM. The challenge of translating ischemic conditioning from animal models to humans: the role of comorbidities. Dis Model Mech 2015; 7:1321-33. [PMID: 25481012 PMCID: PMC4257001 DOI: 10.1242/dmm.016741] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Following a period of ischemia (local restriction of blood supply to a tissue), the restoration of blood supply to the affected area causes significant tissue damage. This is known as ischemia-reperfusion injury (IRI) and is a central pathological mechanism contributing to many common disease states. The medical complications caused by IRI in individuals with cerebrovascular or heart disease are a leading cause of death in developed countries. IRI is also of crucial importance in fields as diverse as solid organ transplantation, acute kidney injury and following major surgery, where post-operative organ dysfunction is a major cause of morbidity and mortality. Given its clinical impact, novel interventions are urgently needed to minimize the effects of IRI, not least to save lives but also to reduce healthcare costs. In this Review, we examine the experimental technique of ischemic conditioning, which entails exposing organs or tissues to brief sub-lethal episodes of ischemia and reperfusion, before, during or after a lethal ischemic insult. This approach has been found to confer profound tissue protection against IRI. We discuss the translation of ischemic conditioning strategies from bench to bedside, and highlight where transition into human clinical studies has been less successful than in animal models, reviewing potential reasons for this. We explore the challenges that preclude more extensive clinical translation of these strategies and emphasize the role that underlying comorbidities have in altering the efficacy of these strategies in improving patient outcomes.
Collapse
Affiliation(s)
- Kieran McCafferty
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK.
| | - Suzanne Forbes
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK
| | - Christoph Thiemermann
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK
| | - Muhammad M Yaqoob
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK
| |
Collapse
|
21
|
Casós K, Pérez ML, Blasco-Lucas A, Ferrer-Curriu G, Gracia-Baena JM, Sureda C, Permanyer E, Igual A, Galiñanes M. Ischemic postconditioning of the isolated human myocardium: Role of the applied protocol. IJC HEART & VASCULATURE 2015; 8:55-61. [PMID: 28785680 PMCID: PMC5497256 DOI: 10.1016/j.ijcha.2015.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/03/2015] [Indexed: 01/06/2023]
Abstract
Background Ischemic postconditioning (IPostC), has been proposed as a useful approach to reduce infarct size in all species, but its clinical utility remains unclear. Objective To investigate the role played by the protocol used on the efficacy of IPostC in protecting the diseased human myocardium. Methods Myocardial atrial samples from patients were subjected to a 90 min ischemia/120 min reoxygenation followed by different IPostC protocols to investigate the role of the time of ischemia (30, 60, 90 and 120 s) and the number of cycles (1, 2, 3 and 4) with 60 and 120 s of total ischemic time. Muscles were also subjected to ischemic preconditioning (IPreC). The release of lactate dehydrogenase (LDH) and the measurement of tetrazolium bromide (MTT) were determined. Results IPostC increased the LDH and decreased the MTT values from those of control, independently of the duration of the conditioning ischemia. LDH and MTT values also worsened by augmenting the number of IPostC cycles whereas they were significantly improved by IPreC. However, analysis of individual results indicated that in approximately 1/3 of the cases IPostC exhibited some degree of protection especially in the presence of increased ischemic injury. Conclusions The present findings show that IPostC of the human myocardium may be influenced by the protocol used and also by the degree of the preceding ischemic injury. IPostC was beneficial in approximately 1/3 of the cases; however in the remaining cases it increased ischemic damage and, therefore, these results raise a word of caution on its broad clinical use.
Collapse
Affiliation(s)
- Kelly Casós
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María-Llanos Pérez
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Arnau Blasco-Lucas
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Ferrer-Curriu
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Manuel Gracia-Baena
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos Sureda
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eduard Permanyer
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Igual
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Galiñanes
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Cyclosporine A reduces microvascular obstruction and preserves left ventricular function deterioration following myocardial ischemia and reperfusion. Basic Res Cardiol 2015; 110:18. [PMID: 25720581 PMCID: PMC4342514 DOI: 10.1007/s00395-015-0475-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 01/31/2015] [Accepted: 02/18/2015] [Indexed: 12/13/2022]
Abstract
Postconditioning and cyclosporine A prevent mitochondrial permeability transition pore opening providing cardioprotection during ischemia/reperfusion. Whether microvascular obstruction is affected by these interventions is largely unknown. Pigs subjected to coronary occlusion for 1 h followed by 3 h of reperfusion were assigned to control (n = 8), postconditioning (n = 9) or cyclosporine A intravenous infusion 10–15 min before the end of ischemia (n = 8). Postconditioning was induced by 8 cycles of repeated 30-s balloon inflation and deflation. After 3 h of reperfusion magnetic resonance imaging, triphenyltetrazolium chloride/Evans blue staining and histopathology were performed. Microvascular obstruction (MVO, percentage of gadolinium-hyperenhanced area) was measured early (3 min) and late (12 min) after contrast injection. Infarct size with double staining was smaller in cyclosporine (46.2 ± 3.1 %, P = 0.016) and postconditioning pigs (47.6 ± 3.9 %, P = 0.008) versus controls (53.8 ± 4.1 %). Late MVO was significantly reduced by cyclosporine (13.9 ± 9.6 %, P = 0.047) but not postconditioning (23.6 ± 11.7 %, P = 0.66) when compared with controls (32.0 ± 16.9 %). Myocardial blood flow in the late MVO was improved with cyclosporine versus controls (0.30 ± 0.06 vs 0.21 ± 0.03 ml/g/min, P = 0.002) and was inversely correlated with late-MVO extent (R2 = 0.93, P < 0.0001). Deterioration of left ventricular ejection fraction (LVEF) between baseline and 3 h of reperfusion was smaller with cyclosporine (−7.9 ± 2.4 %, P = 0.008) but not postconditioning (−12.0 ± 5.5 %, P = 0.22) when compared with controls (−16.4 ± 5.5 %). In the three groups, infarct size (β = −0.69, P < 0.001) and late MVO (β = −0.33, P = 0.02) were independent predictors of LVEF deterioration following ischemia/reperfusion (R2 = 0.73, P < 0.001). Despite both cyclosporine A and postconditioning reduce infarct size, only cyclosporine A infusion had a beneficial effect on microvascular damage and was associated with better preserved LV function when compared with controls.
Collapse
|
23
|
Przyklenk K. Ischaemic conditioning: pitfalls on the path to clinical translation. Br J Pharmacol 2015; 172:1961-73. [PMID: 25560903 DOI: 10.1111/bph.13064] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 12/02/2014] [Accepted: 12/19/2014] [Indexed: 12/21/2022] Open
Abstract
The development of novel adjuvant strategies capable of attenuating myocardial ischaemia-reperfusion injury and reducing infarct size remains a major, unmet clinical need. A wealth of preclinical evidence has established that ischaemic 'conditioning' is profoundly cardioprotective, and has positioned the phenomenon (in particular, the paradigms of postconditioning and remote conditioning) as the most promising and potent candidate for clinical translation identified to date. However, despite this preclinical consensus, current phase II trials have been plagued by heterogeneity, and the outcomes of recent meta-analyses have largely failed to confirm significant benefit. As a result, the path to clinical application has been perceived as 'disappointing' and 'frustrating'. The goal of the current review is to discuss the pitfalls that may be stalling the successful clinical translation of ischaemic conditioning, with an emphasis on concerns regarding: (i) appropriate clinical study design and (ii) the choice of the 'right' preclinical models to facilitate clinical translation.
Collapse
Affiliation(s)
- Karin Przyklenk
- Cardiovascular Research Institute and Departments of Physiology and Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
24
|
Barsukevich V, Basalay M, Sanchez J, Mrochek A, Whittle J, Ackland GL, Gourine AV, Gourine A. Distinct cardioprotective mechanisms of immediate, early and delayed ischaemic postconditioning. Basic Res Cardiol 2014; 110:452. [PMID: 25449894 PMCID: PMC4250560 DOI: 10.1007/s00395-014-0452-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 10/10/2014] [Accepted: 10/24/2014] [Indexed: 12/18/2022]
Abstract
Cardioprotection against ischaemia/reperfusion injury in mice can be achieved by delayed ischaemic postconditioning (IPost) applied as late as 30 min after the onset of reperfusion. We determined the efficacy of delayed IPost in a rat model of myocardial infarction (MI) and investigated potential underlying mechanisms of this phenomenon. Rats were subjected to 20, 30 or 45 min of coronary artery occlusion followed by 120 min of reperfusion (I/R). Immediate and early IPost included six cycles of I/R (10/10 s) applied 10 s or 10 min after reperfusion onset. In the second series of experiments, the rats were subjected to 30 min of coronary occlusion followed by IPost applied 10 s, 10, 30, 45 or 60 min after the onset of reperfusion. Immediate and early IPost (applied 10 s or 10 min of reperfusion) established cardioprotection only when applied after a period of myocardial ischaemia lasting 30 min. Delayed IPost applied after 30 or 45 min of reperfusion reduced infarct sizes by 36 and 41 %, respectively (both P < 0.01). IPost applied 60 min after reperfusion onset was ineffective. Inhibition of RISK pathway (administration of ERK1/2 inhibitor PD-98059 or PI3K inhibitor LY-294002) abolished cardioprotection established by immediate IPost but had no effect on cardioprotection conferred by early IPost. Blockade of SAFE pathway using JAK/STAT inhibitor AG490 had no effect on the immediate or early IPost cardioprotection. Blockade of mitochondrial KATP (mitoKATP) channels (with 5-Hydroxydecanoate) abolished cardioprotection achieved by immediate and early IPost, but had no effect on cardioprotection when IPost was applied 30 or 45 min into the reperfusion period. Immediate IPost increased phosphorylation of PI3K-AKT and ERK1/2. Early or delayed IPost had no effect on phosphorylation of PI3K-AKT, ERK1/2 or STAT3. These data show that in the rat model, delayed IPost confers significant cardioprotection even if applied 45 min after onset of reperfusion. Cardioprotection induced by immediate and early postconditioning involves recruitment of RISK pathway and/or mitoKATP channels, while delayed postconditioning appears to rely on a different mechanism.
Collapse
|
25
|
Wider J, Przyklenk K. Ischemic conditioning: the challenge of protecting the diabetic heart. Cardiovasc Diagn Ther 2014; 4:383-96. [PMID: 25414825 DOI: 10.3978/j.issn.2223-3652.2014.10.05] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
The successful clinical translation of novel therapeutic strategies to attenuate lethal myocardial ischemia-reperfusion injury and limit infarct size has been identified as a major unmet need, and is of particular importance in patients with type-2 diabetes. There is a wealth of preclinical evidence that ischemic conditioning (encompassing the three paradigms of preconditioning, postconditioning and remote conditioning) is profoundly cardioprotective and, via up-regulation of endogenous signaling cascades, renders the heart resistant to infarction. However, current phase II trials aimed at exploiting ischemic conditioning for the clinical treatment of myocardial ischemia-reperfusion injury have yielded mixed results, possibly reflecting the emerging concern that the efficacy of conditioning-induced cardioprotection may be compromised in the diabetic heart. Our goal in this review is to provide a summary of our present understanding of the effect of type-2 diabetes on the infarct-sparing effect of ischemic conditioning, and the challenges of limiting ischemia-reperfusion injury in the diabetic heart.
Collapse
Affiliation(s)
- Joseph Wider
- 1 Cardiovascular Research Institute, 2 Department of Physiology, 3 Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Karin Przyklenk
- 1 Cardiovascular Research Institute, 2 Department of Physiology, 3 Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
26
|
Khan AR, Binabdulhak AA, Alastal Y, Khan S, Faricy-Beredo BM, Luni FK, Lee WM, Khuder S, Tinkel J. Cardioprotective role of ischemic postconditioning in acute myocardial infarction: a systematic review and meta-analysis. Am Heart J 2014; 168:512-521.e4. [PMID: 25262261 DOI: 10.1016/j.ahj.2014.06.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 06/15/2014] [Indexed: 12/24/2022]
Abstract
BACKGROUND Evidence suggests that ischemic postconditioning (IPoC) may reduce the extent of reperfusion injury. We performed a meta-analysis of randomized controlled trials, which compared the role of IPoC during primary percutaneous coronary intervention (PCI) to PCI alone (control group) in ST-segment elevation myocardial infarction. METHODS Several databases were searched, which yielded 19 studies. The outcomes of interest were measures of myocardial damage (serum cardiac enzymes and infarct size by imaging) and left ventricular function (left ventricular ejection fraction and wall motion score index). Mean difference (MD) and standardized mean difference (SMD) were used to assess the treatment effect. An inverse variance method was used to pool data into a random-effects model. RESULTS Ischemic postconditioning demonstrated a decrease in serum cardiac enzymes (SMD -0.48, 95% CI -0.92 to -0.05, I(2) = 92%), reduction in infarct size by imaging (SMD -0.30, 95% CI -0.58 to -0.01, I(2) = 80%), wall motion score index (MD -0.19, 95% CI -0.29 to -0.09, I(2) = 44%), and showed improvement in left ventricular ejection fraction (IPoC 52 ± 0.4, control 49.7 ± 0.4) (MD 2.78, 95% CI 0.66-4.91, I(2) = 69%). All included studies were limited by high risk of performance and publication bias. CONCLUSIONS Ischemic postconditioning during PCI in ST-segment elevation myocardial infarction appears to be superior to PCI alone in reduction of both myocardial injury or damage and improvement in global and regional left ventricular function. The effect seems to be more pronounced when a greater myocardial area is at risk. Given the limitations of the current available evidence, additional data from large randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Abdur Rahman Khan
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH
| | - Aref A Binabdulhak
- Department of Internal Medicine, University of Missouri - Kansas City, Kansas, MO
| | - Yaseen Alastal
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH
| | - Sobia Khan
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH
| | | | - Faraz Khan Luni
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH
| | - Wade M Lee
- Mulford Health Science Library - University of Toledo, Toledo, OH
| | - Sadik Khuder
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH
| | - Jodi Tinkel
- Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH.
| |
Collapse
|