51
|
Hu Z, Chen M, Zhang P, Liu J, Abbott GW. Remote ischemic preconditioning differentially attenuates post-ischemic cardiac arrhythmia in streptozotocin-induced diabetic versus nondiabetic rats. Cardiovasc Diabetol 2017; 16:57. [PMID: 28446231 PMCID: PMC5406986 DOI: 10.1186/s12933-017-0537-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/19/2017] [Indexed: 02/05/2023] Open
Abstract
Background Sudden cardiac death (SCD), a leading cause of global mortality, most commonly arises from a substrate of cardiac ischemia, but requires an additional trigger. Diabetes mellitus (DM) predisposes to SCD even after adjusting for other DM-linked cardiovascular pathology such as coronary artery disease. We previously showed that remote liver ischemia preconditioning (RLIPC) is highly protective against cardiac ischemia reperfusion injury (IRI) linked ventricular arrhythmias and myocardial infarction, via induction of the cardioprotective RISK pathway, and specifically, inhibitory phosphorylation of GSK-3β (Ser 9). Methods We evaluated the impact of acute streptozotocin-induced DM on coronary artery ligation IRI-linked ventricular arrhythmogenesis and RLIPC therapy in rats. Results Post-IRI arrhythmia induction was similar in nondiabetic and DM rats, but, unexpectedly, DM rats exhibited lower incidence of SCD during reperfusion (41 vs. 100%), suggesting uncontrolled hyperglycemia does not acutely predispose to SCD. RLIPC was highly effective in both nondiabetic and DM rats at reducing incidence and duration of, and increasing latency to, all classes of ventricular tachyarrhythmias. In contrast, atrioventricular block (AVB) was highly responsive to RLIPC in nondiabetic rats (incidence reduced from 72 to 18%) but unresponsive in DM rats. RISK pathway induction was similar in nondiabetic and DM rats, thus not explaining the DM-specific resistance of AVB to therapy. Conclusions Our findings uncover important acute DM-specific differences in responsiveness to remote preconditioning for ventricular tachyarrhythmias versus AVB, which may have clinical significance given that AVB is a malignant arrhythmia twofold more common in human diabetics than nondiabetics, and correlated to plasma glucose levels >10 mmol/L.
Collapse
Affiliation(s)
- Zhaoyang Hu
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Mou Chen
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Zhang
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Liu
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Dept. of Pharmacology and Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
52
|
Menting TP, Wever KE, Ozdemir‐van Brunschot DMD, Van der Vliet DJA, Rovers MM, Warle MC. Ischaemic preconditioning for the reduction of renal ischaemia reperfusion injury. Cochrane Database Syst Rev 2017; 3:CD010777. [PMID: 28258686 PMCID: PMC6464274 DOI: 10.1002/14651858.cd010777.pub2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Ischaemia reperfusion injury can lead to kidney dysfunction or failure. Ischaemic preconditioning is a short period of deprivation of blood supply to particular organs or tissue, followed by a period of reperfusion. It has the potential to protect kidneys from ischaemia reperfusion injury. OBJECTIVES This review aimed to look at the benefits and harms of local and remote ischaemic preconditioning to reduce ischaemia and reperfusion injury among people with renal ischaemia reperfusion injury. SEARCH METHODS We searched Cochrane Kidney and Transplant's Specialised Register to 5 August 2016 through contact with the Information Specialist using search terms relevant to this review. SELECTION CRITERIA We included all randomised controlled trials measuring kidney function and the role of ischaemic preconditioning in patients undergoing a surgical intervention that induces kidney injury. Kidney transplantation studies were excluded. DATA COLLECTION AND ANALYSIS Studies were assessed for eligibility and quality; data were extracted by two independent authors. We collected basic study characteristics: type of surgery, remote ischaemic preconditioning protocol, type of anaesthesia. We collected primary outcome measurements: serum creatinine and adverse effects to remote ischaemic preconditioning and secondary outcome measurements: acute kidney injury, need for dialysis, neutrophil gelatinase-associated lipocalin, hospital stay and mortality. Summary estimates of effect were obtained using a random-effects model, and results were expressed as risk ratios (RR) and their 95% confidence intervals (CI) for dichotomous outcomes, and mean difference (MD) and 95% CI for continuous outcomes. MAIN RESULTS We included 28 studies which randomised a total of 6851 patients. Risk of bias assessment indicated unclear to low risk of bias for most studies. For consistency regarding the direction of effects, continuous outcomes with negative values, and dichotomous outcomes with values less than one favour remote ischaemic preconditioning. Based on high quality evidence, remote ischaemic preconditioning made little or no difference to the reduction of serum creatinine levels at postoperative days one (14 studies, 1022 participants: MD -0.02 mg/dL, 95% CI -0.05 to 0.02; I2 = 21%), two (9 studies, 770 participants: MD -0.04 mg/dL, 95% CI -0.09 to 0.02; I2 = 31%), and three (6 studies, 417 participants: MD -0.05 mg/dL, 95% CI -0.19 to 0.10; I2 = 68%) compared to control.Serious adverse events occurred in four patients receiving remote ischaemic preconditioning by iliac clamping. It is uncertain whether remote ischaemic preconditioning by cuff inflation leads to increased adverse effects compared to control because the certainty of the evidence is low (15 studies, 3993 participants: RR 3.47, 95% CI 0.55 to 21.76; I2 = 0%); only two of 15 studies reported any adverse effects (6/1999 in the remote ischaemic preconditioning group and 1/1994 in the control group), the remaining 13 studies stated no adverse effects were observed in either group.Compared to control, remote ischaemic preconditioning made little or no difference to the need for dialysis (13 studies, 2417 participants: RR 0.85, 95% CI 0.37 to 1.94; I2 = 60%; moderate quality evidence), length of hospital stay (8 studies, 920 participants: MD 0.17 days, 95% CI -0.46 to 0.80; I2 = 49%, high quality evidence), or all-cause mortality (24 studies, 4931 participants: RR 0.86, 95% CI 0.54 to 1.37; I2 = 0%, high quality evidence).Remote ischaemic preconditioning may have slightly improved the incidence of acute kidney injury using either the AKIN (8 studies, 2364 participants: RR 0.76, 95% CI 0.57 to 1.00; I2 = 61%, high quality evidence) or RIFLE criteria (3 studies, 1586 participants: RR 0.91, 95% CI 0.75 to 1.12; I2 = 0%, moderate quality evidence). AUTHORS' CONCLUSIONS Remote ischaemic preconditioning by cuff inflation appears to be a safe method, and probably leads to little or no difference in serum creatinine, adverse effects, need for dialysis, length of hospital stay, death and in the incidence of acute kidney injury. Overall we had moderate-high certainty evidence however the available data does not confirm the efficacy of remote ischaemic preconditioning in reducing renal ischaemia reperfusion injury in patients undergoing major cardiac and vascular surgery in which renal ischaemia reperfusion injury may occur.
Collapse
Affiliation(s)
- Theo P Menting
- Radboud University Nijmegen Medical CentreDepartment of SurgeryGeert Grooteplein Zuid 10NijmegenGelderlandNetherlands6525 GA
| | - Kimberley E Wever
- Radboud University Nijmegen Medical CentreDepartment of SurgeryGeert Grooteplein Zuid 10NijmegenGelderlandNetherlands6525 GA
| | - Denise MD Ozdemir‐van Brunschot
- Radboud University Nijmegen Medical CentreDepartment of SurgeryGeert Grooteplein Zuid 10NijmegenGelderlandNetherlands6525 GA
| | - Daan JA Van der Vliet
- Radboud University Nijmegen Medical CentreDepartment of SurgeryGeert Grooteplein Zuid 10NijmegenGelderlandNetherlands6525 GA
| | - Maroeska M Rovers
- Radboud University Nijmegen Medical CentreDepartment of Operating RoomsHp 630, route 631PO Box 9101NijmegenNetherlands6500 HB
| | - Michiel C Warle
- Radboud University Nijmegen Medical CentreDepartment of SurgeryGeert Grooteplein Zuid 10NijmegenGelderlandNetherlands6525 GA
| | | |
Collapse
|
53
|
Gedik N, Maciel L, Schulte C, Skyschally A, Heusch G, Kleinbongard P. Cardiomyocyte mitochondria as targets of humoral factors released by remote ischemic preconditioning. Arch Med Sci 2017; 13:448-458. [PMID: 28261301 PMCID: PMC5332452 DOI: 10.5114/aoms.2016.61789] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 06/30/2016] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Remote ischemic preconditioning (RIPC) reduces myocardial infarct size, and protection can be transferred with plasma to other individuals, even across species. Mitochondria are the end-effectors of cardioprotection by local ischemic conditioning maneuvers. We have now analyzed mitochondrial function in response to RIPC. MATERIAL AND METHODS Plasma from pigs undergoing placebo or RIPC (infarct size reduction by 67% in RIPC pigs compared to placebo) was transferred to isolated perfused rat hearts subjected to 30 min global ischemia followed by 120 min reperfusion for infarct size measurement. Additional experiments were terminated at 10 min reperfusion to isolate mitochondria for functional measurements. Effects of RIPC pig plasma were compared to local ischemic preconditioning (IPC) or to infusion of tumor necrosis factor α (TNF-α). RESULTS Ischemia/reperfusion (I/R) induced an infarct of 41 ±2% of total ventricular mass. Placebo pig plasma did not affect infarct size (38 ±1, p = 0.13). The RIPC pig plasma reduced infarct size (27 ±2, p < 0.001), as did IPC (20 ±1, p < 0.001) and TNF-α (28 ±2, p < 0.001). Associated with cardioprotection, reductions of mitochondrial adenosine diphosphate (ADP)-stimulated respiration, adenosine triphosphate (ATP) production and calcium retention capacity (CRC) by I/R and placebo pig plasma were prevented by RIPC pig plasma, as they were by IPC and TNF-α. Mitochondrial reactive oxygen species production (nmol H2O2/100 µg protein) induced by I/R (272 ±34) was comparable in response to placebo pig plasma (234 ±28, p = 0.37) and was reduced by RIPC pig plasma (83 ±15, p < 0.001) as well as by IPC (78 ±21, p < 0.001) and TNF-α (125 ±42, p = 0.002). CONCLUSIONS In rat myocardium, mitochondria are an intracellular target of protection induced by humoral factors retrieved from pigs undergoing RIPC.
Collapse
Affiliation(s)
- Nilguen Gedik
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen, Medical School, Essen, Germany
| | - Leonardo Maciel
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen, Medical School, Essen, Germany
- Laboratory of Cardiac Electrophysiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Christiane Schulte
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen, Medical School, Essen, Germany
| | - Andreas Skyschally
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen, Medical School, Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen, Medical School, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen, Medical School, Essen, Germany
| |
Collapse
|
54
|
Wang S, He W, Wang C. MiR-23a Regulates the Vasculogenesis of Coronary Artery Disease by Targeting Epidermal Growth Factor Receptor. Cardiovasc Ther 2017; 34:199-208. [PMID: 27085964 DOI: 10.1111/1755-5922.12187] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Circulating microRNAs (miRNAs) in patient body fluids have recently been considered to hold the potential of being novel disease biomarkers and drug targets. We aimed to investigate the correlation between the levels of circulating miR-23a and the expression of epidermal growth factor receptor (EGFR) in the pathogenesis of patients with coronary heart disease to further explore the mechanism involved in its vasculogenesis. METHOD Three different cohorts, including 13 acute myocardial infarction (AMI) patients, 176 angina pectoris patients, and 127 control subjects, were enrolled to investigate the expression levels of circulating miR-23a in patients with myocardial ischemia and also the relationship between plasma miR-23a and severity of coronary stenosis. Plasma miR-23a levels of participants were examined by real-time quantitative PCR. Simultaneously, plasma cardiac troponin I (cTnI) concentrations were measured by ELISAs. We further detected the correlation of miR-23a and EGFR by molecular and animal assays. RESULT MiR-23a was enriched in not only diseased endothelial progenitor cells (EPCs) but also in the plasma of patients with coronary artery disease (CAD). Besides, we found out miR-23a was able to suppress EGFR expression and EPC activities. Reporter assays confirmed the direct binding and repression of miR-23a to the 3'-UTR of EGFR mRNA. Knockdown of miR-23a not only restored EGFR levels and angiogenic activities of diseased EPCs in vitro, but further promoted blood flow recovery in ischemic limbs of mice. CONCLUSION Circulating miR-23a may be a new biomarker for CAD and as a potential diagnostic tool. And increased miR-23a level may be used to predict the presence and severity of coronary lesions in patients with CAD.
Collapse
Affiliation(s)
- Shixi Wang
- Department of Cardiology, Central Hospital of Zaozhuang Mining Group, Zaozhuang, Shandong, China
| | - Weidong He
- Department of Nutritional, Affiliated Hospital, Qingdao University Medical College, Qingdao, Shandong, China
| | - Caijin Wang
- Department of Cardiology, Institute of Traditional Chinese Medicine, Liaocheng, Shandong, China
| |
Collapse
|
55
|
Cardioprotection by remote ischemic conditioning and its signal transduction. Pflugers Arch 2016; 469:159-181. [DOI: 10.1007/s00424-016-1922-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/23/2022]
|
56
|
Biglino G, Caputo M, Rajakaruna C, Angelini G, van Rooij E, Emanueli C. Modulating microRNAs in cardiac surgery patients: Novel therapeutic opportunities? Pharmacol Ther 2016; 170:192-204. [PMID: 27902930 DOI: 10.1016/j.pharmthera.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review focuses on microRNAs (miRs) in cardiac surgery, where they are emerging as potential targets for therapeutic intervention as well as novel clinical biomarkers. Identification of the up/down-regulation of specific miRs in defined groups of cardiac surgery patients can lead to the development of novel strategies for targeted treatment in order to maximise therapeutic results and minimise acute, delayed or chronic complications. MiRs could also be involved in determining the outcome independently of complications, for example in relation to myocardial perfusion and fibrosis. Because of their relevance in disease, their known sequence and pharmacological properties, miRs are attractive candidates for therapeutic manipulation. Pharmacological inhibition of individual miRs can be achieved by modified antisense oligonucleotides, referred to as antimiRs, while miR replacement can be achieved by miR mimics to increase the level of a specific miR. MiR mimics can restore the function of a lost or down-regulated miR, while antimiRs can inhibit the levels of disease-driving or aberrantly expressed miRs, thus de-repressing the expression of mRNAs targeted by the miR. The main delivery methods for miR therapeutics involve lipid-based vehicles, viral systems, cationic polymers, and intravenous or local injection of an antagomiR. Local delivery is particularly desirable for miR therapeutics and options include the development of devices specific for local delivery, light-induced antimiR, and vesicle-encapsulated miRs serving as therapeutic delivery agents able to improve intracellular uptake. Here, we discuss the potential therapeutic use of miRNAs in the context of cardiac surgery.
Collapse
Affiliation(s)
| | - Massimo Caputo
- Bristol Heart Institute, University of Bristol, Bristol, UK; RUSH University Medical Center, Chicago, IL, USA
| | - Cha Rajakaruna
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | | | | | - Costanza Emanueli
- Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
57
|
Lotfi AS, Eftekhari H, Atreya AR, Kashikar A, Sivalingam SK, Giannoni M, Visintainer P, Engelman D. Randomized controlled trial of remote ischemic preconditioning and atrial fibrillation in patients undergoing cardiac surgery. World J Cardiol 2016; 8:615-622. [PMID: 27847563 PMCID: PMC5088368 DOI: 10.4330/wjc.v8.i10.615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/26/2016] [Accepted: 08/29/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To study whether remote ischemic preconditioning (RIPC) has an impact on clinical outcomes, such as post-operative atrial fibrillation (POAF).
METHODS This was a prospective, single-center, single-blinded, randomized controlled study. One hundred and two patients were randomized to receive RIPC (3 cycles of 5 min ischemia and 5 min reperfusion in the upper arm after induction of anesthesia) or no RIPC (control). Primary outcome was POAF lasting for five minutes or longer during the first seven days after surgery. Secondary outcomes included length of hospital stay, incidence of inpatient mortality, myocardial infarction, and stroke.
RESULTS POAF occurred at a rate of 54% in the RIPC group and 41.2% in the control group (P = 0.23). No statistically significant differences were noted in secondary outcomes between the two groups.
CONCLUSION This is the first study in the United States to suggest that RIPC does not reduce POAF in patients with elective or urgent cardiac surgery. There were no differences in adverse effects in either group. Further studies are required to assess the relationship between RIPC and POAF.
Collapse
|
58
|
Kosiuk J, Milani R, Ueberham L, Uhe T, Stegmann C, Hindricks G, Bollmann A. Effect of remote ischemic preconditioning on electrophysiological and biomolecular parameters in nonvalvular paroxysmal atrial fibrillation (RIPPAF study): Rationale and study design of a randomized, controlled clinical trial. Clin Cardiol 2016; 39:631-635. [PMID: 27775830 DOI: 10.1002/clc.22584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 11/09/2022] Open
Abstract
Remote ischemic preconditioning (RIPC) has been studied in models of different cardiovascular entities. Recently, a beneficial effect of RIPC on incidence of atrial fibrillation (AF) in postsurgical patients has been suggested. However, the potential impact of RIPC on electrophysiological- and thrombogenesis-related parameters in the setting of paroxysmal nonvalvular AF has not been investigated. The aim of the study is to answer the following questions: (1) Does RIPC have impact on inducibility of AF in patients with known paroxysmal AF? If yes, what are the direct electrophysiological mechanisms of this phenomenon, and could RIPC be implemented to reduce AF burden? (2) Does RIPC have the potential to minimize thrombogenic effects of simulated episodes of AF? If so, what are inhibited components of thrombogenesis and can this be used to reduce thromboembolic risk related to paroxysmal AF? The presented study is a 2-arm, randomized, placebo-controlled, double-blinded, single-center trial in a cohort of 146 patients with paroxysmal AF referred for AF ablation in sinus rhythm. The study will collect electrophysiological data such as variability of P-wave morphology, atrial refractory period, conduction times, and inducibility/sustainability of AF. Furthermore, AF-induced prothrombotic processes will be analyzed by quantification of platelet aggregates, analysis of platelet function, and measurement of thrombogenesis-related plasma markers. Moreover, the study will provide a unique bio-database for further analysis of molecular and genetic mechanisms responsible for observed results.
Collapse
Affiliation(s)
- Jedrzej Kosiuk
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| | - Romina Milani
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| | - Laura Ueberham
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| | - Tobias Uhe
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| | - Clara Stegmann
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| | - Gerhard Hindricks
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| | - Andreas Bollmann
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Leipzig, Germany
| |
Collapse
|
59
|
Abstract
The mortality from acute myocardial infarction (AMI) remains significant, and the prevalence of post-myocardial infarction heart failure is increasing. Therefore, cardioprotection beyond timely reperfusion is needed. Conditioning procedures are the most powerful cardioprotective interventions in animal experiments. However, ischemic preconditioning cannot be used to reduce infarct size in patients with AMI because its occurrence is not predictable; several studies in patients undergoing surgical coronary revascularization report reduced release of creatine kinase and troponin. Ischemic postconditioning reduces infarct size in most, but not all, studies in patients undergoing interventional reperfusion of AMI, but may require direct stenting and exclusion of patients with >6 hours of symptom onset to protect. Remote ischemic conditioning reduces infarct size in patients undergoing interventional reperfusion of AMI, elective percutaneous or surgical coronary revascularization, and other cardiovascular surgery in many, but not in all, studies. Adequate dose-finding phase II studies do not exist. There are only 2 phase III trials, both on remote ischemic conditioning in patients undergoing cardiovascular surgery, both with neutral results in terms of infarct size and clinical outcome, but also both with major problems in trial design. We discuss the difficulties in translation of cardioprotection from animal experiments and proof-of-concept trials to clinical practice. Given that most studies on ischemic postconditioning and all studies on remote ischemic preconditioning in patients with AMI reported reduced infarct size, it would be premature to give up on cardioprotection.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology (G.H.) and Clinic for Cardiology (T.R.), West German Heart and Vascular Center, University School of Medicine Essen, Essen, Germany
| | - Tienush Rassaf
- From the Institute for Pathophysiology (G.H.) and Clinic for Cardiology (T.R.), West German Heart and Vascular Center, University School of Medicine Essen, Essen, Germany
| |
Collapse
|
60
|
Han R, Liu X, Yin X, Zheng M, Sun K, Liu X, Tian Y, Yang X. Effect of remote ischemic preconditioning on myocardial injury and inflammatory response induced by ablation for atrial fibrillation: A randomized controlled trial. Int J Cardiol 2016; 222:396-400. [PMID: 27505322 DOI: 10.1016/j.ijcard.2016.07.229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/29/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) has been suggested to reduce postoperative release of cardiac and inflammatory markers in patients undergoing cardiac surgery. This study aimed to evaluate the effect of RIPC on nonischemic myocardial damage and inflammatory response in patients undergoing radiofrequency catheter ablation for paroxysmal atrial fibrillation (AF). METHODS Seventy-two patients with drug-refractory paroxysmal AF undergoing radiofrequency catheter ablation were randomized into RIPC or control groups. RIPC (intermittent arm ischemia through four cycles of 5-min inflation and 5-min deflation of a blood-pressure cuff) was performed once daily on 2 consecutive days before the ablation and immediately before ablation. Cardiac troponin-I (cTnI), high-sensitive C-reactive protein (hs-CRP), and interleukin (IL)-6 levels were measured before RIPC/sham RIPC, after the ablation, and 24 and 72h later. The early recurrence of atrial fibrillation (ERAF) in the two groups was observed over the subsequent 3months. RESULTS Radiofrequency ablation resulted in a significant rise in cTnI, hs-CRP, and IL-6 in both groups, which persisted for 72h. The RIPC group showed a lower increase in cTnI (P<0.001), hs-CRP (P=0.003), and IL-6 (P=0.008) than the control and tended to have a lower risk of ERAF (hazard ratio [HR]=0.77, 95% confidence interval [CI]: 0.32-1.88). CONCLUSIONS These results show that RIPC before ablation for paroxysmal AF significantly reduces the increase in cTnI, hs-CRP, and IL-6 associated with the procedure and results in a lower risk of ERAF. These findings suggest that RIPC could provide cardioprotection against nonischemic myocardial damage.
Collapse
Affiliation(s)
- Ruijuan Han
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaoqing Liu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - XianDong Yin
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Meili Zheng
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Kai Sun
- Department of Radiology, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Xi-Cheng District, Beijing 100037, China
| | - Xingpeng Liu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Ying Tian
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xinchun Yang
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
61
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac-specific miRNA in cardiogenesis, heart function, and cardiac pathology (with focus on myocardial infarction). J Mol Cell Cardiol 2016; 94:107-121. [PMID: 27056419 DOI: 10.1016/j.yjmcc.2016.03.015] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/09/2016] [Accepted: 03/24/2016] [Indexed: 12/21/2022]
Abstract
Cardiac miRNAs (miR-1, miR133a, miR-208a/b, and miR-499) are abundantly expressed in the myocardium. They play a central role in cardiogenesis, heart function and pathology. While miR-1 and miR-133a predominantly control early stages of cardiogenesis supporting commitment of cardiac-specific muscle lineage from embryonic stem cells and mesodermal precursors, miR-208 and miR-499 are involved in the late cardiogenic stages mediating differentiation of cardioblasts to cardiomyocytes and fast/slow muscle fiber specification. In the heart, miR-1/133a control cardiac conductance and automaticity by regulating all phases of the cardiac action potential. miR-208/499 located in introns of the heavy chain myosin genes regulate expression of sarcomeric contractile proteins. In cardiac pathology including myocardial infarction (MI), expression of cardiac miRNAs is markedly altered that leads to deleterious effects associated with heart wounding, arrhythmia, increased apoptosis, fibrosis, hypertrophy, and tissue remodeling. In acute MI, circulating levels of cardiac miRNAs are significantly elevated making them to be a promising diagnostic marker for early diagnosis of acute MI. Great cardiospecific capacity of these miRNAs is very helpful for enhancing regenerative properties and survival of stem cell and cardiac progenitor transplants and for reprogramming of mature non-cardiac cells to cardiomyocytes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Department of Biophysics, Biological Faculty, Moscow State University, Moscow 119991, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
62
|
Apoptosis-related microRNA changes in the right atrium induced by remote ischemic perconditioning during valve replacement surgery. Sci Rep 2016; 6:18959. [PMID: 26738985 PMCID: PMC4704063 DOI: 10.1038/srep18959] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 11/13/2015] [Indexed: 01/05/2023] Open
Abstract
We previously found that remote ischemic perconditioning (RIPerc) was effective in attenuating myocardial injury during cardiac surgery. Given that microRNAs (miRs) act as an important player in ischemic/reperfusion (I/R) injury and apoptosis, this study aimed to investigate whether RIPerc reduces apoptosis in atrial myocardium and which apoptosis-related miRs are involved during valve replacement surgery. Here, we demonstrated that RIPerc inhibited apoptosis in atrial myocardium during cardiac ischemia and that 17 miRs showed at least a 1.5-fold change in expression after ischemia. Of the 17 miRs, 9 miRs, including miR-1, miR-21, miR-24, and miR-195, which are related to apoptosis, exhibited different expression patterns in the RIPerc group compared with the control. Using qRT-PCR and Western blotting, we demonstrated that miR-1 and miR-195 were downregulated and that their common putative target gene Bcl-2 was upregulated in the RIPerc group. However, the differences in miR-21 and miR-24 expression, together with programmed cell death 4 (PDCD4), which is the target gene of miR-21, were not significant. These findings provide some insight into the role of miRs in the cardioprotective effects induced by RIPerc.
Collapse
|
63
|
|
64
|
Sharma V, Marsh R, Cunniffe B, Cardinale M, Yellon DM, Davidson SM. From Protecting the Heart to Improving Athletic Performance - the Benefits of Local and Remote Ischaemic Preconditioning. Cardiovasc Drugs Ther 2015; 29:573-588. [PMID: 26477661 PMCID: PMC4674524 DOI: 10.1007/s10557-015-6621-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Remote Ischemic Preconditioning (RIPC) is a non-invasive cardioprotective intervention that involves brief cycles of limb ischemia and reperfusion. This is typically delivered by inflating and deflating a blood pressure cuff on one or more limb(s) for several cycles, each inflation-deflation being 3-5 min in duration. RIPC has shown potential for protecting the heart and other organs from injury due to lethal ischemia and reperfusion injury, in a variety of clinical settings. The mechanisms underlying RIPC are under intense investigation but are just beginning to be deciphered. Emerging evidence suggests that RIPC has the potential to improve exercise performance, via both local and remote mechanisms. This review discusses the clinical studies that have investigated the role of RIPC in cardioprotection as well as those studying its applicability in improving athletic performance, while examining the potential mechanisms involved.
Collapse
Affiliation(s)
- Vikram Sharma
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Reuben Marsh
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Brian Cunniffe
- English institute of Sport, Bisham, Marlow, UK
- Institute of Sport, Exercise and Health, UCL, London, UK
| | - Marco Cardinale
- Institute of Sport, Exercise and Health, UCL, London, UK
- Aspire Academy, Doha, Qatar
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
65
|
Benstoem C, Stoppe C, Liakopoulos OJ, Meybohm P, Clayton TC, Yellon DM, Hausenloy DJ, Goetzenich A. Remote ischaemic preconditioning for coronary artery bypass grafting. Cochrane Database Syst Rev 2015. [PMCID: PMC4676907 DOI: 10.1002/14651858.cd011719.pub2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This is the protocol for a review and there is no abstract. The objectives are as follows: To assess the benefits and harms of remote ischaemic preconditioning in patients undergoing coronary artery bypass grafting, with or without valve surgery.
Collapse
Affiliation(s)
- Carina Benstoem
- Department of Cardiothoracic Surgery, University Hospital AachenAachen, Germany
- Contact address: Carina Benstoem, Department of Cardiothoracic Surgery, University Hospital Aachen, Pauwelsstrasse 30, Aachen, 52074, Germany.
| | - Christian Stoppe
- Department of Anesthesiology, University Hospital AachenAachen, Germany
| | - Oliver J Liakopoulos
- Department of Cardiothoracic Surgery, Heart Center, University of CologneCologne, Germany
| | - Patrick Meybohm
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital FrankfurtFrankfurt am Main, Germany
| | - Tim C Clayton
- Department of Medical Statistics, London School of Hygiene & Tropical MedicineLondon, UK
| | - Derek M Yellon
- Department of Medicine, University College London Hospital and Medical SchoolLondon, UK
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College LondonLondon, UK
| | - Andreas Goetzenich
- Department of Cardiothoracic Surgery, University Hospital AachenAachen, Germany
- Contact address: Carina Benstoem, Department of Cardiothoracic Surgery, University Hospital Aachen, Pauwelsstrasse 30, Aachen, 52074, Germany.
| |
Collapse
|
66
|
Tao H, Shi KH, Yang JJ, Li J. Epigenetic mechanisms in atrial fibrillation: New insights and future directions. Trends Cardiovasc Med 2015; 26:306-18. [PMID: 26475117 DOI: 10.1016/j.tcm.2015.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 08/23/2015] [Accepted: 08/28/2015] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. AF is a complex disease that results from genetic and environmental factors and their interactions. In recent years, numerous studies have shown that epigenetic mechanisms significantly participate in AF pathogenesis. Even though a poor understanding of the molecular and electrophysiologic mechanisms of AF, accumulated evidence has suggested that the relevance of epigenetic changes in the development of AF. The aim of this review is to describe the present knowledge about the epigenetic regulatory features significantly participates in AF, and look ahead on new perspectives of epigenetic mechanisms research. Epigenetic regulatory features such as DNA methylation, histone modification, and microRNA influence gene expression by epigenetic mechanisms and by directly binding to various factor response elements in the target gene promoters. Given the role of epigenetic alterations in regulating genes, there is potential for the integration of factors-induced epigenetic alterations as informative factors in the risk assessment process. In this review, new insight into the epigenetic mechanisms in AF pathogenesis is discussed, with special emphasis on DNA methylation, histone modification, and microRNA. Further studies are needed to reveal the potential targets of epigenetic mechanisms, and it can be developed as a therapeutic target for AF.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China; Cardiovascular Research Center, Anhui Medical University, Hefei, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China; Cardiovascular Research Center, Anhui Medical University, Hefei, China.
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
67
|
Cardiac over-expression of microRNA-1 induces impairment of cognition in mice. Neuroscience 2015; 299:66-78. [PMID: 25943483 DOI: 10.1016/j.neuroscience.2015.04.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 11/23/2022]
Abstract
Large cohort studies have revealed a close relationship between cognitive impairment and cardiovascular diseases, although the mechanism underlying this relationship remains incompletely understood. In this study, using a transgenic (Tg) mouse model of cardiac-specific over-expression of microRNA-1-2 (miR-1-2), we observed that microRNA-1 (miR-1) levels were increased not only in the heart but also in the hippocampus and blood, whereas its levels did not change in the skeletal muscle of Tg mice compared with age-matched wild-type (WT) mice. Six-month-old Tg mice showed cognitive impairment compared with age-matched WT mice, as assessed using the Morris Water Maze test. The brain-derived neurotrophic factor (BDNF) level and cyclic AMP-responsive element-binding protein (CREB) phosphorylation were also significantly reduced in the hippocampi of the Tg mice, as evaluated by Western blot. Further examination showed that BDNF protein expression was down- or up-regulated by miR-1 over-expression or inhibition, respectively, and was unchanged by binding site mutations or miRNA-masks for the 3'UTR of Bdnf, indicating that this gene is a potential target of miR-1. Knockdown of miR-1 by hippocampal stereotaxic injection of an anti-miR-1 oligonucleotide fragment carried by a lentivirus vector (lenti-pre-AMO-miR-1) led to up-regulation of BDNF expression and prevented the reduction in cognitive performance in the Tg mice without affecting cardiac function. Our findings demonstrate that cardiac over-expression of miR-1 also induces behavioral abnormalities that may be associated, at least in part, with the down-regulation of BDNF expression in the hippocampus. This study definitely contributes to the understanding of the relationship between cardiovascular disease and cognitive impairment.
Collapse
|
68
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
69
|
Abstract
Mitochondrial dysfunction has been shown to be involved in the pathophysiology of arrhythmia, not only in inherited cardiomyopathy due to specific mutations in the mitochondrial DNA but also in acquired cardiomyopathy such as ischemic or diabetic cardiomyopathy. This article briefly discusses the basics of mitochondrial physiology and details the mechanisms generating arrhythmias due to mitochondrial dysfunction. The clinical spectrum of inherited and acquired cardiomyopathies associated with mitochondrial dysfunction is discussed followed by general aspects of the management of mitochondrial cardiomyopathy and related arrhythmia.
Collapse
Affiliation(s)
- David Montaigne
- Lille University, Inserm U1011, European Genomic Institute for Diabetes, Place de Verdun-amphi J&K, Lille F-59045, France; Institut Pasteur de Lille, Boulevard Louis XV, Lille F-59019, France; Cardiovascular Explorations Department, University Hospital of Lille, Lille F-59000, France.
| | - Anju Duva Pentiah
- Cardiovascular Explorations Department, University Hospital of Lille, Lille F-59000, France; Division of Cardiomyopathy, Department of Cardiology, University Hospital of Lille, Rue du Pr Laguesse, Lille F-59000, France
| |
Collapse
|
70
|
Hudetz JA, Patterson KM, Iqbal Z, Gandhi SD, Pagel PS. Remote Ischemic Preconditioning Prevents Deterioration of Short-Term Postoperative Cognitive Function After Cardiac Surgery Using Cardiopulmonary Bypass: Results of a Pilot Investigation. J Cardiothorac Vasc Anesth 2015; 29:382-8. [DOI: 10.1053/j.jvca.2014.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Indexed: 11/11/2022]
|
71
|
Oba T, Yasukawa H, Nagata T, Kyogoku S, Minami T, Nishihara M, Ohshima H, Mawatari K, Nohara S, Takahashi J, Sugi Y, Igata S, Iwamoto Y, Kai H, Matsuoka H, Takano M, Aoki H, Fukumoto Y, Imaizumi T. Renal Nerve-Mediated Erythropoietin Release Confers Cardioprotection During Remote Ischemic Preconditioning. Circ J 2015; 79:1557-67. [PMID: 25833080 DOI: 10.1253/circj.cj-14-1171] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) induced by transient limb ischemia is a powerful innate mechanism of cardioprotection against ischemia. Several described mechanisms explain how RIPC may act through neural pathways or humoral factors; however, the mechanistic pathway linking the remote organ to the heart has not yet been fully elucidated. This study aimed to investigate the mechanisms underlying the RIPC-induced production of Janus kinase (JAK)-signal transducer and activator of the transcription (STAT)-activating cytokines and cardioprotection by using mouse and human models of RIPC. METHODS AND RESULTS Screened circulating cardioprotective JAK-STAT-activating cytokines in mice unexpectedly revealed increased serum erythropoietin (EPO) levels after RIP induced by transient ischemia. In mice, RIPC rapidly upregulated EPO mRNA and its main transcriptional factor, hypoxia-inducible factor-1α (HIF1α), in the kidney. Laser Doppler blood flowmetry revealed a prompt reduction of renal blood flow (RBF) after RIPC. RIPC activated cardioprotective signaling pathways and the anti-apoptotic Bcl-xL pathway in the heart, and reduced infarct size. In mice, these effects were abolished by administration of an EPO-neutralizing antibody. Renal nerve denervation also abolished RIPC-induced RBF reduction, EPO production, and cardioprotection. In humans, transient limb ischemia of the upper arm reduced RBF and increased serum EPO levels. CONCLUSIONS Based on the present data, we propose a novel RIPC mechanism in which inhibition of infarct size by RIPC is produced through the renal nerve-mediated reduction of RBF associated with activation of the HIF1α-EPO pathway.
Collapse
Affiliation(s)
- Toyoharu Oba
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Myrmel T. Translational research: Sounds intriguing, but can at times be a frustrating endeavor. How can we improve our methodology? SCAND CARDIOVASC J 2015; 49:115-6. [PMID: 25752845 DOI: 10.3109/14017431.2015.1026388] [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: 11/13/2022]
Affiliation(s)
- Truls Myrmel
- Department of Cardiothoracic and Vascular Surgery, University Hospital North Norway and the Institute of Clinical Medicine, University of Tromsø, the Arctic University of Norway , Norway
| |
Collapse
|
73
|
Montaigne D, Maréchal X, Lacroix D, Staels B. From cardiac mitochondrial dysfunction to clinical arrhythmias. Int J Cardiol 2015; 184:597-599. [PMID: 25769006 DOI: 10.1016/j.ijcard.2015.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/01/2015] [Indexed: 02/02/2023]
Affiliation(s)
- David Montaigne
- Université Lille 2, F-59000 Lille, France; Inserm, U1011, F-59000 Lille, France; EGID, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France; Service d'Explorations Fonctionnelles CardioVasculaires, University Hospital of Lille, France.
| | - Xavier Maréchal
- Université Lille 2, F-59000 Lille, France; Inserm, U1011, F-59000 Lille, France; EGID, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| | - Dominique Lacroix
- Clinique de Cardiologie, Service de Rythmologie, University Hospital of Lille, France
| | - Bart Staels
- Université Lille 2, F-59000 Lille, France; Inserm, U1011, F-59000 Lille, France; EGID, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| |
Collapse
|
74
|
Krogstad LEB, Slagsvold KH, Wahba A. Remote ischemic preconditioning and incidence of postoperative atrial fibrillation. SCAND CARDIOVASC J 2015; 49:117-22. [PMID: 25613907 DOI: 10.3109/14017431.2015.1010565] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Although remote ischemic preconditioning (RIPC) has shown favorable effects on ischemia-reperfusion injury, much remains unknown of its mechanisms and clinical significance. We hypothesized that RIPC would reduce the incidence of postoperative atrial fibrillation (POAF) following coronary artery bypass graft (CABG) surgery. In addition, we investigated whether RIPC could induce alterations of circulating microRNA in blood plasma. DESIGN This is a single-center, double-blind, randomized controlled trial. 92 adult patients referred for first-time isolated CABG surgery were randomly assigned to either RIPC (n = 45) or control (n = 47). The RIPC-stimulus comprised three 5-min cycles of upper arm ischemia, induced by inflating a blood pressure cuff to 200 mmHg, with an intervening 5 min reperfusion. Heart rhythm was assessed by telemetry. MicroRNA expression was assessed in plasma by real-time polymerase chain reaction. RESULTS Of the 92 patients included in the study, 27 patients developed POAF (29%). 17 of these patients belonged to the RIPC group (38%), and 10 to the control group (21%). There were no significant alterations of microRNA expression. CONCLUSIONS We did not observe a reduced incidence of POAF by RIPC before CABG surgery. Larger multi-center studies may be necessary to further clarify this issue.
Collapse
|
75
|
Remote ischemic conditioning and cardioprotection: a systematic review and meta-analysis of randomized clinical trials. Basic Res Cardiol 2015; 110:11. [DOI: 10.1007/s00395-015-0467-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/25/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022]
|
76
|
Slagsvold KH, Wahba A. Molecular mechanisms in remote ischaemic preconditioning: from the research lab to clinical routine? Eur J Cardiothorac Surg 2015; 48:738. [PMID: 25646397 DOI: 10.1093/ejcts/ezu556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Katrine H Slagsvold
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway Department of Cardiothoracic Surgery, St Olav's University Hospital, Trondheim, Norway
| | - Alexander Wahba
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway Department of Cardiothoracic Surgery, St Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
77
|
Heusch G, Bøtker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. J Am Coll Cardiol 2015; 65:177-95. [PMID: 25593060 PMCID: PMC4297315 DOI: 10.1016/j.jacc.2014.10.031] [Citation(s) in RCA: 490] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022]
Abstract
In remote ischemic conditioning (RIC), brief, reversible episodes of ischemia with reperfusion in one vascular bed, tissue, or organ confer a global protective phenotype and render remote tissues and organs resistant to ischemia/reperfusion injury. The peripheral stimulus can be chemical, mechanical, or electrical and involves activation of peripheral sensory nerves. The signal transfer to the heart or other organs is through neuronal and humoral communications. Protection can be transferred, even across species, with plasma-derived dialysate and involves nitric oxide, stromal derived factor-1α, microribonucleic acid-144, but also other, not yet identified factors. Intracardiac signal transduction involves: adenosine, bradykinin, cytokines, and chemokines, which activate specific receptors; intracellular kinases; and mitochondrial function. RIC by repeated brief inflation/deflation of a blood pressure cuff protects against endothelial dysfunction and myocardial injury in percutaneous coronary interventions, coronary artery bypass grafting, and reperfused acute myocardial infarction. RIC is safe and effective, noninvasive, easily feasible, and inexpensive.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany.
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Karin Przyklenk
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Andrew Redington
- Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Derek Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| |
Collapse
|
78
|
MiR-214 regulates the pathogenesis of patients with coronary artery disease by targeting VEGF. Mol Cell Biochem 2015; 402:111-22. [PMID: 25575606 DOI: 10.1007/s11010-014-2319-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/23/2014] [Indexed: 12/11/2022]
Abstract
Circulating microRNAs (miRNAs) in patient body fluids have recently been considered to hold the potential of being novel disease biomarkers and drug targets. We aimed to investigate the correlation between the levels of circulating miR-214 and the expression of vascular endothelial growth factor (VEGF) in the pathogenesis of coronary heart disease patients to further explore the mechanism involved in the vasculogenesis. Three different cohorts, including 13 acute myocardial infarction patients, 176 angina pectoris patients, and 127 control subjects, were enrolled to investigate the expression levels of circulating miR-214 in patients with myocardial ischemia and also the relationship between plasma miR-214 and severity of coronary stenosis. Plasma miR-214 levels of participants were examined by real-time quantitative PCR. Simultaneously, plasma cardiac troponin I concentrations were measured by ELISA assays. We further detected the correlation of miR-214 and VEGF by molecular and animal assays. MiR-214 was enriched in not only diseased endothelial progenitor cells (EPCs) but also the plasma of coronary artery disease (CAD) patients. Besides, we found out miR-214 was able to suppress VEGF expression and EPC activities. Reporter assays confirmed the direct binding and repression of miR-214 to the 39-UTR of VEGF mRNA. Knockdown of miR-214 not only restored VEGF levels and angiogenic activities of diseased EPCs in vitro, but also further promoted blood flow recovery in ischemic limbs of mice. Circulating miR-214 may be a new biomarker for CAD and as a potential diagnostic tool. And increased miR-214 level may be used to predict the presence and severity of coronary lesions in CAD patients.
Collapse
|
79
|
Remote ischemic preconditioning for myocardial protection: update on mechanisms and clinical relevance. Mol Cell Biochem 2015; 402:41-9. [PMID: 25552250 DOI: 10.1007/s11010-014-2312-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/20/2014] [Indexed: 02/07/2023]
Abstract
Ischemic heart disease is the leading cause of death for both men and women worldwide, accruing 7.4 million deaths in 2012. There has been a continued search for better cardioprotective modalities that would reduce myocardial ischemia-reperfusion injury. Among these attempts, a more convenient model of ischemic preconditioning, known as remote ischemic preconditioning (RIPC) was first introduced in 1993 by Przyklenk and colleagues who reported that brief regional occlusion-reperfusion episodes in one vascular bed of the heart render protection to remote myocardial tissue. Subsequently, major advances in myocardial RIPC came with the use of skeletal muscle as the ischemic stimulus. To date, numerous studies have revealed that RIPC applied to the kidney, liver, mesentery, and skeletal muscle, have all exhibited cardioprotective effects. The main purpose of this review article is to summarize the new advances in understanding the molecular mechanisms of RIPC during the past 5 years, including those related to capsaicin-activated C sensory fibers, hypoxia-inducible factor 1α, connexin 43, extracellular vesicles, microRNA-144, microRNA-1, and nitrite. In addition, we have discussed results from several recent human clinical trials with RIPC. Taken together, the emerging clinical evidence supports the concept that the effectiveness of RIPC paired with its low-cost and non-invasive features makes it an ideal treatment before reperfusion after sustained ischemia. More carefully designed studies are warranted to fully exploit the clinical benefits of RIPC and its potential implications in patients with cardiovascular disease.
Collapse
|
80
|
FERKO M, KANCIROVÁ I, JAŠOVÁ M, ČARNICKÁ S, MURÁRIKOVÁ M, WACZULÍKOVÁ I, SUMBALOVÁ Z, KUCHARSKÁ J, ULIČNÁ O, RAVINGEROVÁ T, ZIEGELHÖFFER A. Remote Ischemic Preconditioning of the Heart: Protective Responses in Functional and Biophysical Properties of Cardiac Mitochondria. Physiol Res 2014; 63:S469-78. [DOI: 10.33549/physiolres.932933] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Remote ischemic preconditioning (RIP)-induced protection of myocardial energetics was well documented on the level of tissue, but data concerning the involvement of mitochondria were missing. We aimed at the identification of changes in membrane properties and respiratory functions induced in rat heart mitochondria by RIP. Experiments were performed on 46 male Wistar rats divided into control and RIP-treated groups of 21 animals each. Blood flow in the occluded area was recorded by MRI angiography in four animals. RIP protocol comprised of three successive 5-min occlusions each followed by 5-min reperfusions of descending branches of the right hind limb femoral artery. The efficacy of RIP was evaluated as the extent of RIP-induced protection against damage to the functions of mitochondria isolated by differential centrifugation after 30-min global ischemia followed by 40-min reperfusion of the hearts in Langendorff mode. Assessments: mitochondrial membrane fluidity with a fluorescent probe DPH, CoQ9 and CoQ10 with HPLC, mitochondrial respiration with the Oxygraph-2k (Oroboros). Results revealed that RIP was affecting the mitochondria. The immediate protection conferred by RIP involves beneficial and prognostically significant effects: a total elimination of ischemia/reperfusion-induced depression of mitochondrial membrane fluidity and a trend for better preservation of mitochondrial state 3 respiration.
Collapse
Affiliation(s)
- M. FERKO
- Institute for Heart Research, Centre of Excellence SAS NOREG, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Ai F, Chen M, Li W, Yang Y, Xu G, Gui F, Liu Z, Bai X, Chen Z. Protective role of Klotho on cardiomyocytes upon hypoxia/reoxygenation via downregulation of Akt and FOXO1 phosphorylation. Mol Med Rep 2014; 11:2013-9. [PMID: 25377663 DOI: 10.3892/mmr.2014.2899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/05/2014] [Indexed: 11/05/2022] Open
Abstract
Klotho is a novel anti-aging hormone involved in human coronary artery disease. The present study aimed to detect the effects and mechanism of Klotho on cardiomyocytes in a hypoxia/reoxygenation (H/R) model in vitro. Neonatal Sprague-Dawley rat cardiomyocytes were randomly distributed into experimental groups as follows: Control group; H/R group, 4‑h hypoxia followed by 3‑h reoxygenation; and H/R+Klotho group, incubated with 0.1, 0.2 or 0.4 µg/ml Klotho protein for 16 h and then subjected to 4‑h hypoxia/3‑h reoxygenation. In order to evaluate cardiomyocyte damage, cell viability and lactate dehydrogenase (LDH) levels were measured. Cell apoptosis was measured by flow cytometry. The 2',7'-dichlorofluorescein diacetate reagent was used to estimate the intracellular generation of reactive oxygen species (ROS). Immunofluorescence staining was used to test whether Klotho induced decreased nuclear translocation of forkhead box protein O1 (FOXO1). Western blot analysis was performed to detect protein levels of FOXO1, phospho-FOXO1, Akt, phospho-Akt and superoxide dismutase 2 (SOD2). Cell viability was significantly decreased, levels of LDH in the cardiomyocyte culture medium were significantly increased and the apoptotic rate was enhanced in the H/R group when compared with those of the control group. Compared with the H/R group, cell viability of the H/R+Klotho groups was significantly higher (P<0.05). Treatment with Klotho protein resulted in a significant resistance of cardiomyocytes to apoptosis and the release of LDH was decreased. Intracellular ROS levels in the H/R group were significantly elevated above those of the control group (P<0.05). Following treatment with Klotho, intracellular ROS levels were significantly decreased compared with those of the H/R group (P<0.05). Western blot analysis confirmed that Klotho protein treatment increased FOXO1 levels in the nucleus and decreased FOXO1 levels in the cytoplasm. Furthermore, exogenous Klotho protein promoted translocation of FOXO1 from cytoplasm to nucleus. In addition, the administration of Klotho protein suppressed phosphorylation of FOXO1 and Akt, and markedly increased the protein expression levels of SOD2. In conclusion, treatment with Klotho protein had beneficial effects on cardiomyocytes undergoing H/R injury. The mechanism of this effect may be associated with suppressed apoptosis of cardiomyocytes, inhibition of phosphorylation of FOXO1 and Akt as well as suppression of cytoplasm transfer of FOXO1.
Collapse
Affiliation(s)
- Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Wei Li
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yang Yang
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Guizhong Xu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Feng Gui
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhenxing Liu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Xiangyan Bai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
82
|
Calway T, Kim GH. Harnessing the Therapeutic Potential of MicroRNAs for Cardiovascular Disease. J Cardiovasc Pharmacol Ther 2014; 20:131-43. [PMID: 25261390 DOI: 10.1177/1074248414552902] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are one of the most common causes of death in humans and are responsible for billions of dollars in health care expenditures. As the molecular basis of cardiac diseases continues to be explored, there remains the hope for identification of more effective therapeutics. MicroRNAs (miRNAs) are recognized as important regulators of numerous biological pathways and stress responses, including those found in cardiovascular diseases. MicroRNA signatures of cardiovascular diseases can provide targets for miRNA adjustment and offer the possibility of changing gene and protein expression to treat certain pathologies. These adjustments can be conferred using advances in oligonucleotide delivery methods, which can target single miRNAs, families of miRNAs, and certain tissue types. In this review, we will discuss the use of miRNAs in vivo and recent advances in their use for cardiovascular disease in mammalian models.
Collapse
Affiliation(s)
- Tyler Calway
- Institute for Cardiovascular Research, University of Chicago, Chicago, IL, USA
| | - Gene H Kim
- Institute for Cardiovascular Research, University of Chicago, Chicago, IL, USA
| |
Collapse
|
83
|
Li J, Rohailla S, Gelber N, Rutka J, Sabah N, Gladstone RA, Wei C, Hu P, Kharbanda RK, Redington AN. MicroRNA-144 is a circulating effector of remote ischemic preconditioning. Basic Res Cardiol 2014; 109:423. [PMID: 25060662 DOI: 10.1007/s00395-014-0423-z] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/11/2014] [Accepted: 06/23/2014] [Indexed: 02/06/2023]
Abstract
Remote ischemic preconditioning (rIPC) induced by cycles of transient limb ischemia and reperfusion is a powerful cardioprotective strategy with additional pleiotropic effects. However, our understanding of its underlying mediators and mechanisms remains incomplete. We examined the role of miR-144 in the cardioprotection induced by rIPC. Microarray studies first established that rIPC increases, and IR injury decreases miR-144 levels in mouse myocardium, the latter being rescued by both rIPC and intravenous administration of miR-144. Going along with this systemic treatment with miR-144 increased P-Akt, P-GSK3β and P-p44/42 MAPK, decreased p-mTOR level and induced autophagy signaling, and induced early and delayed cardioprotection with improved functional recovery and reduction in infarct size similar to that achieved by rIPC. Conversely, systemic administration of a specific antisense oligonucleotide reduced myocardial levels of miR-144 and abrogated cardioprotection by rIPC. We then showed that rIPC increases plasma miR-144 levels in mice and humans, but there was no change in plasma microparticle (50-400 nM) numbers or their miR-144 content. However, there was an almost fourfold increase in miR-144 precursor in the exosome pellet, and a significant increase in miR-144 levels in exosome-poor serum which, in turn, was associated with increased levels of the miR carriage protein Argonaute-2. Systemic release of microRNA 144 plays a pivotal role in the cardioprotection induced by rIPC. Future studies should assess the potential for plasma miR-144 as a biomarker of the effectiveness of rIPC induced by limb ischemia, and whether miR-144 itself may represent a novel therapy to reduce clinical ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jing Li
- Division of Cardiology, Labatt Family Heart Center, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X 8, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Hale A, Lee C, Annis S, Min PK, Pande R, Creager MA, Julian CG, Moore LG, Mitsialis SA, Hwang SJ, Kourembanas S, Chan SY. An Argonaute 2 switch regulates circulating miR-210 to coordinate hypoxic adaptation across cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2528-42. [PMID: 24983771 DOI: 10.1016/j.bbamcr.2014.06.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 06/05/2014] [Accepted: 06/23/2014] [Indexed: 10/25/2022]
Abstract
Complex organisms may coordinate molecular responses to hypoxia by specialized avenues of communication across multiple tissues, but these mechanisms are poorly understood. Plasma-based, extracellular microRNAs have been described, yet their regulation and biological functions in hypoxia remain enigmatic. We found a unique pattern of release of the hypoxia-inducible microRNA-210 (miR-210) from hypoxic and reoxygenated cells. This microRNA is also elevated in human plasma in physiologic and pathologic conditions of altered oxygen demand and delivery. Released miR-210 can be delivered to recipient cells, and the suppression of its direct target ISCU and mitochondrial metabolism is primarily evident in hypoxia. To regulate these hypoxia-specific actions, prolyl-hydroxylation of Argonaute 2 acts as a molecular switch that reciprocally modulates miR-210 release and intracellular activity in source cells as well as regulates intracellular activity in recipient cells after miR-210 delivery. Therefore, Argonaute 2-dependent control of released miR-210 represents a unique communication system that integrates the hypoxic response across anatomically distinct cells, preventing unnecessary activity of delivered miR-210 in normoxia while still preparing recipient tissues for incipient hypoxic stress and accelerating adaptation.
Collapse
Affiliation(s)
- Andrew Hale
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Changjin Lee
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, 02215, USA
| | - Sofia Annis
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Pil-Ki Min
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Cardiology Division, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Reena Pande
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Mark A Creager
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Colleen G Julian
- Department of Emergency Medicine, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Lorna G Moore
- Department of Medicine, and Obstetrics-Gynecology, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, 02215, USA
| | - Sarah J Hwang
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, 02215, USA
| | - Stephen Y Chan
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
85
|
Przyklenk K, Sanderson TH, Hüttemann M. Clinical benefits of remote ischemic preconditioning: new insights...and new questions. Circ Res 2014; 114:748-50. [PMID: 24577960 DOI: 10.1161/circresaha.114.303331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Karin Przyklenk
- From the Cardiovascular Research Institute (K.P., T.H.S., M.H.), Center for Molecular Medicine and Genetics (K.P., M.H.), Department of Physiology (K.P., T.H.S.), and Department of Emergency Medicine (K.P., T.H.S.), Wayne State University School of Medicine, Detroit, MI
| | | | | |
Collapse
|
86
|
Santulli G, Iaccarino G, De Luca N, Trimarco B, Condorelli G. Atrial fibrillation and microRNAs. Front Physiol 2014; 5:15. [PMID: 24478726 PMCID: PMC3900852 DOI: 10.3389/fphys.2014.00015] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/08/2014] [Indexed: 12/17/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia, especially in the elderly, and has a significant genetic component. Recently, several independent investigators have demonstrated a functional role for small non-coding RNAs (microRNAs) in the pathophysiology of this cardiac arrhythmia. This report represents a systematic and updated appraisal of the main studies that established a mechanistic association between specific microRNAs and AF, focusing both on the regulation of electrical and structural remodeling of cardiac tissue.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Advanced Biomedical Sciences, "Federico II" University Hospital Naples, Italy ; Department of Translational Medical Sciences, "Federico II" University Hospital Naples, Italy ; Columbia University Medical Center, College of Physicians & Surgeons, New York Presbyterian Hospital - Manhattan New York, NY, USA
| | - Guido Iaccarino
- Department of Medicine and Surgery, University of Salerno Salerno, Italy ; IRCCS "Multimedica," Milano, Italy
| | - Nicola De Luca
- Department of Translational Medical Sciences, "Federico II" University Hospital Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University Hospital Naples, Italy
| | - Gianluigi Condorelli
- Humanitas Clinical and Research Center Rozzano (Milan), Italy ; University of Milan Milan, Italy
| |
Collapse
|