51
|
Mi L, Zhang N, Wan J, Cheng M, Liao J, Zheng X. Remote ischemic post‑conditioning alleviates ischemia/reperfusion‑induced intestinal injury via the ERK signaling pathway‑mediated RAGE/HMGB axis. Mol Med Rep 2021; 24:773. [PMID: 34490475 PMCID: PMC8441982 DOI: 10.3892/mmr.2021.12413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal ischemia reperfusion (I/R) injury is a tissue and organ injury that frequently occurs during surgery and significantly contributes to the pathological processes of severe infection, injury, shock, cardiopulmonary insufficiency and other diseases. However, the mechanism of intestinal I/R injury remains to be elucidated. A mouse model of intestinal I/R injury was successfully established and the model mice were treated with remote ischemic post‑conditioning (RIPOC) and/or an ERK inhibitor (CC‑90003), respectively. Histopathological changes of the intestinal mucosa were determined by hematoxylin and eosin staining. In addition, the levels of high‑mobility group box 1 (HMGB1) and receptor for advanced glycation end products (RAGE) expression were confirmed by reverse transcription‑quantitative polymerase chain reaction, western blotting and immunohistochemistry assays. The levels of antioxidants, oxidative stress markers (8‑OHdG) and interleukin 1 family members were evaluated by ELISA assays and the levels of NF‑κB pathway proteins were analyzed by western blotting. The data demonstrated that RIPOC could attenuate the histopathological features of intestinal mucosa in the intestinal I/R‑injury mouse models via the ERK pathway. It was also revealed that HMGB1 and RAGE expression in the mouse models could be markedly reduced by RIPOC (P<0.05) and that these reductions were associated with inhibition of the ERK pathway. Furthermore, it was demonstrated that RIPOC produced significant antioxidant and anti‑inflammatory effects following an intestinal I/R injury and that these effects were mediated via the ERK pathway (P<0.05). In addition, RIPOC was demonstrated to suppress the NF‑κB (p65)/NLR family pyrin domain containing 3 (NLRP3) inflammatory pathways in the intestinal I/R injury mouse models via the ERK pathway. The findings of the present study demonstrated that RIPOC helped to protect mice with an intestinal I/R injury by downregulating the ERK pathway.
Collapse
Affiliation(s)
- Lei Mi
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Nan Zhang
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Jiyun Wan
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Ming Cheng
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Jianping Liao
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xiao Zheng
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
52
|
Jarosz J, Trybulski R, Krzysztofik M, Tsoukos A, Filip-Stachnik A, Zajac A, Bogdanis GC, Wilk M. The Effects of Ischemia During Rest Intervals on Bar Velocity in the Bench Press Exercise With Different External Loads. Front Physiol 2021; 12:715096. [PMID: 34447318 PMCID: PMC8383203 DOI: 10.3389/fphys.2021.715096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/22/2021] [Indexed: 11/13/2022] Open
Abstract
The main aim of the present study was to evaluate the acute effects of ischemia used during rest periods on bar velocity changes during the bench press exercise at progressive loads, from 20 to 90% of 1RM. Ten healthy resistance trained men volunteered for the study (age = 26.3 ± 4.7 years; body mass = 89.8 ± 6.3 kg; bench press 1RM = 142.5 ± 16.9 kg; training experience = 7.8 ± 2.7 years). During the experimental sessions the subjects performed the bench press exercise under two different conditions, in a randomized and counterbalanced order: (a) ischemia condition, with ischemia applied before the first set and during every rest periods between sets, and (b) control condition where no ischemia was applied. During each experimental session eight sets of the bench press exercise were performed, against loads starting from 20 to 90% 1RM, increased progressively by 10% in each subsequent set. A 3-min rest interval between sets was used. For ischemia condition the cuffs was applied 3 min before the first set and during every rest period between sets. Ischemia was released during exercise. The cuff pressure was set to ∼80% of full arterial occlusion pressure. The two-way repeated measures ANOVA showed a statistically significant interaction effect for peak bar velocity (p = 0.04) and for mean bar velocity (p = 0.01). There was also a statistically significant main effect of condition for peak bar velocity (p < 0.01) but not for mean bar velocity (p = 0.25). The post hoc analysis for interaction showed significantly higher peak bar velocity for the ischemia condition compared to control at a load of 20% 1RM (p = 0.007) and at a load of 50% 1RM (p = 0.006). The results of the present study indicate that ischemia used before each set even for a brief duration of <3 min, has positive effects on peak bar velocity at light loads, but it is insufficient to induce such effect on higher loads.
Collapse
Affiliation(s)
- Jakub Jarosz
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Robert Trybulski
- Department of Medical Sciences, The Wojciech Korfanty School of Economics, Katowice, Poland.,Provita Zory Medical Center, Zory, Poland
| | - Michał Krzysztofik
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Athanasios Tsoukos
- School of Physical Education and Sport Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Aleksandra Filip-Stachnik
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Adam Zajac
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Gregory C Bogdanis
- School of Physical Education and Sport Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Michal Wilk
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| |
Collapse
|
53
|
Krag AE, Hvas CL, Kiil BJ, Hvas AM. Effect of Remote Ischemic Conditioning on Bleeding Complications in Surgery: A Systematic Review and Meta-Analysis. Semin Thromb Hemost 2021; 48:229-239. [PMID: 34428800 DOI: 10.1055/s-0041-1732468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Remote ischemic conditioning (RIC) is administered with an inflatable tourniquet by inducing brief, alternating cycles of limb ischemia and reperfusion. RIC possibly impacts the hemostatic system, and the intervention has been tested as protective therapy against ischemia-reperfusion injury and thrombotic complications in cardiac surgery and other surgical procedures. In the present systematic review, we aimed to investigate the effect of RIC on intraoperative and postoperative bleeding complications in meta-analyses of randomized controlled trials including adult patients undergoing surgery. A systematic search was performed on November 7, 2020 in PubMed, Embase, and the Cochrane Central Register of Controlled Trials. Randomized controlled trials comparing RIC versus no RIC in adult patients undergoing surgery that reported bleeding outcomes in English publications were included. Effect estimates with 95% confidence intervals were calculated using the random-effects model for intraoperative and postoperative bleeding outcomes. Thirty-two randomized controlled trials with 3,804 patients were eligible for inclusion. RIC did not affect intraoperative bleeding volume (nine trials; 392 RIC patients, 399 controls) with the effect estimate -0.95 [-9.90; 7.99] mL (p = 0.83). RIC significantly reduced postoperative drainage volume (seven trials; 367 RIC patients, 365 controls) with mean difference -83.6 [-134.9; -32.4] mL (p = 0.001). The risk of re-operation for bleeding was reduced in the RIC group (16 trials; 838 RIC patients, 839 controls), albeit not significantly, with the relative risk 0.65 [0.39; 1.09] (p = 0.10). In conclusion, RIC reduced postoperative bleeding measured by postoperative drainage volume in this meta-analysis of adult patients undergoing surgery.
Collapse
Affiliation(s)
- Andreas E Krag
- Thrombosis and Hemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Plastic and Breast Surgery, Aarhus University Hospital, Denmark
| | - Christine L Hvas
- Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Denmark
| | - Birgitte J Kiil
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Denmark
| | - Anne-Mette Hvas
- Thrombosis and Hemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
54
|
Bainey KR, Zheng Y, Coulden R, Sonnex E, Thompson R, Mei J, Bastiany A, Welsh R. Remote ischaemic conditioning in ST elevation myocardial infarction: a registry-based randomised trial. Heart 2021; 108:703-709. [PMID: 34417205 DOI: 10.1136/heartjnl-2021-319455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/29/2021] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Remote ischaemic conditioning (RIC) has been tested as a possible strategy for mitigating reperfusion injury in ST elevation myocardial infarction (STEMI) with primary percutaneous coronary intervention (PPCI). However, surrogate outcomes have shown inconsistent effects with lack of clinical correlation. METHODS We performed a registry-based randomised study of patients with STEMI allocated to RIC (4 cycles of blood pressure cuff inflation to 200 mm Hg for 5 min of ischaemia followed by 5 min of reperfusion) or standard of care (SOC) during PPCI. We examined the associations of RIC on core laboratory measurements of myocardial perfusion, infarct size (IS), left ventricular (LV) performance and clinical outcomes. RESULTS A total of 252 patients were enrolled. The median age was 61 (IQR: 55-70) years and 72.8% were male. Sum ST segment deviation resolution ≥50% was similar between RIC and SOC (65.2% vs 55.7%, p=0.269). In those with 3-day cardiovascular MRI (n=88), no difference in median (25th, 75th percentiles) IS (14.9% (4.5%, 23.1%) vs 16.1% (3.3%, 22.0%), p=0.980), LV dimensions (LV end-diastolic volume index: 78.7 (71.1, 91.2) mL/m2 vs 79.9 (71.2, 88.8) mL/m2, p=0.630; LV end-systolic volume index: 48.8 (35.7, 51.4) mL/m2 vs 37.9 (31.8, 47.5) mL/m2, p=0.551) or ejection fraction (50.0% (41.0%-55.0%) vs 50.0% (43.0%-56.0%), p=0.554) was demonstrated. Similar results were observed with 90-day cardiovascular MRI. At 1 year, the clinical composite of death, congestive heart failure, cardiogenic shock and recurrent myocardial infarction was similar in RIC and SOC (21.7% vs 13.3%, p=0.110). CONCLUSIONS In a contemporary registry-based randomised study of patients with STEMI undergoing PPCI, adjunctive therapy with RIC did not improve myocardial perfusion, reduce IS or alter LV performance. Consequently, there was no difference in clinical outcomes within 1 year. TRIAL REGISTRATION NUMBER NCT03930589.
Collapse
Affiliation(s)
- Kevin R Bainey
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada .,Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Yinggan Zheng
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Richard Coulden
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Radiology and Diagnostic Imaging, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Emer Sonnex
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Radiology and Diagnostic Imaging, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Richard Thompson
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Junyi Mei
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Alexandra Bastiany
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Robert Welsh
- Divison of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| |
Collapse
|
55
|
The Effects of Ischemic Preconditioning Supplementation on Endothelial Function: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6690691. [PMID: 34349827 PMCID: PMC8328691 DOI: 10.1155/2021/6690691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/03/2020] [Accepted: 07/16/2021] [Indexed: 01/01/2023]
Abstract
Objective Ischemic preconditioning (IPC) has gradually been promoted in clinical practice to lower the risk of cardiovascular surgery and postoperative complications. We investigated the role of IPC on vascular endothelial function and the relationship between IPC, flow-mediated dilation (FMD), and brachial artery diameter (BAD). Methods Systematic searches were conducted in PubMed, Medline, Cochrane Library, Embase, and Scopus databases from their inception to March 20, 2020. This research included randomized controlled trials (RCTs) with adults, and the values of FMD and BAD were considered as the primary outcomes. Ten studies comprising 292 participants were included in the meta-analysis. Results Regarding FMD, we observed beneficial effects of IPC on endothelial function (standardized mean difference (SMD): 1.82; 95% confidence interval (CI): 0.64, 3.01; p < 0.001; I 2 = 89.9%). However, the available evidence did not indicate that IPC affected BAD (SMD: 0.08; 95% CI: -0.03, 0.18; p > 0.05; I 2 = 76.5%). Conclusions Our meta-analysis indicated a significant effect of IPC on the endothelial function of the blood vessels, affecting FMD but not BAD.
Collapse
|
56
|
Reducing Cardiac Injury during ST-Elevation Myocardial Infarction: A Reasoned Approach to a Multitarget Therapeutic Strategy. J Clin Med 2021; 10:jcm10132968. [PMID: 34279451 PMCID: PMC8268641 DOI: 10.3390/jcm10132968] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/22/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The significant reduction in ‘ischemic time’ through capillary diffusion of primary percutaneous intervention (pPCI) has rendered myocardial-ischemia reperfusion injury (MIRI) prevention a major issue in order to improve the prognosis of ST elevation myocardial infarction (STEMI) patients. In fact, while the ischemic damage increases with the severity and the duration of blood flow reduction, reperfusion injury reaches its maximum with a moderate amount of ischemic injury. MIRI leads to the development of post-STEMI left ventricular remodeling (post-STEMI LVR), thereby increasing the risk of arrhythmias and heart failure. Single pharmacological and mechanical interventions have shown some benefits, but have not satisfactorily reduced mortality. Therefore, a multitarget therapeutic strategy is needed, but no univocal indications have come from the clinical trials performed so far. On the basis of the results of the consistent clinical studies analyzed in this review, we try to design a randomized clinical trial aimed at evaluating the effects of a reasoned multitarget therapeutic strategy on the prevention of post-STEMI LVR. In fact, we believe that the correct timing of pharmacological and mechanical intervention application, according to their specific ability to interfere with survival pathways, may significantly reduce the incidence of post-STEMI LVR and thus improve patient prognosis.
Collapse
|
57
|
Wang H, Zheng Z, Zhang N, Zhou Y, Jin S. Regular transient limb ischemia protects endothelial function against hypercholesterolemic damage in rabbits. Sci Prog 2021; 104:368504211036858. [PMID: 34351826 PMCID: PMC10358469 DOI: 10.1177/00368504211036858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Regular transient limb ischemia (RTLI) can prevent atherosclerosis in hypercholesterolemic rabbits. As endothelial dysfunction is the initial factor leading to atherosclerosis, we investigated the effect of RTLI on endothelial function in hypercholesterolemic rabbits. We randomly allocated 15 New Zealand white rabbits to three groups, five animals per group: the hypercholesterolemic group (Group H), the sham RTLI group (Group S), and the RTLI group (Group L). All rabbits received hypercholesterolemic fodder daily. No intervention was performed on the rabbits in Group H. Rabbits in Group S were kept in hutches, with a deflated cuff applied to their left hind limb for 60 min every day. For rabbits in Group L, RTLI (six cycles of 5-min ischemia and 5-min reperfusion of the left hind limb) was applied once daily for 12 weeks. At the end of week 12, a segment of the abdominal aorta was isolated from each rabbit for in vitro measurement of the endothelium-dependent vasodilation (EDV) response to different concentrations of acetylcholine and the endothelium-independent vasodilation (EIV) response to sodium nitroprusside. The EDV response was significantly higher in Group L than in Groups S and H (p < 0.05), with no significant difference between Groups S and H (p > 0.05). There was no difference in the EIV response among the three groups. RTLI could improve the EDV response, protecting endothelial function against hypercholesterolemic damage.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Anesthesia, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Zhinan Zheng
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nanrong Zhang
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Zhou
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sanqing Jin
- Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
58
|
Slysz JT, Burr JF. Ischemic Preconditioning: Modulating Pain Sensitivity and Exercise Performance. Front Physiol 2021; 12:696488. [PMID: 34239452 PMCID: PMC8258159 DOI: 10.3389/fphys.2021.696488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/01/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose The purpose of this study was to examine whether an individual’s IPC-mediated change in cold pain sensitivity is associated with the same individual’s IPC-mediated change in exercise performance. Methods Thirteen individuals (8 males; 5 females, 27 ± 7 years, 55 ± 5 ml.kgs–1.min–1) underwent two separate cold-water immersion tests: with preceding IPC treatment and without. In addition, each participant undertook two separate 5-km cycling time trials: with preceding IPC treatment and without. Pearson correlation coefficients were used to assess the relationship between an individual’s change in cold-water pain sensitivity following IPC with their change in 5-km time trial performance following IPC. Results During the cold-water immersion test, pain intensity increased over time (p < 0.001) but did not change with IPC (p = 0.96). However, IPC significantly reduced the total time spent under pain (−9 ± 7 s; p = 0.001) during the cold-water immersion test. No relationship was found between an individual’s change in time under pain (r = −0.2, p = 0.6) or pain intensity (r = −0.3, p = 0.3) following IPC and their change in performance following IPC. Conclusion These findings suggest that IPC can modulate sensitivity to a painful stimulus, but this altered sensitivity does not explain the ergogenic efficacy of IPC on 5-km cycling performance.
Collapse
Affiliation(s)
- Joshua T Slysz
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Jamie F Burr
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
59
|
Myocardial remote ischemic preconditioning: from cell biology to clinical application. Mol Cell Biochem 2021; 476:3857-3867. [PMID: 34125317 DOI: 10.1007/s11010-021-04192-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/26/2021] [Indexed: 12/25/2022]
Abstract
Remote ischemic preconditioning (rIPC) is a cardioprotective phenomenon where brief periods of ischemia followed by reperfusion of one organ/tissue can confer subsequent protection against ischemia/reperfusion injury in other organs, such as the heart. It involves activation of humoral, neural or systemic communication pathways inducing different intracellular signals in the heart. The main purpose of this review is to summarize the possible mechanisms involved in the rIPC cardioprotection, and to describe recent clinical trials to establish the efficacy of these strategies in cardioprotection from lethal ischemia/reperfusion injury. In this sense, certain factors weaken the subcellular mechanisms of rIPC in patients, such as age, comorbidities, medication, and anesthetic protocol, which could explain the heterogeneity of results in some clinical trials. For these reasons, further studies, carefully designed, are necessary to develop a clearer understanding of the pathways and mechanism of early and late rIPC. An understanding of the pathways is important for translation to patients.
Collapse
|
60
|
Remote ischemic preconditioning improves tissue oxygenation in a porcine model of controlled hemorrhage without fluid resuscitation. Sci Rep 2021; 11:10808. [PMID: 34031524 PMCID: PMC8144617 DOI: 10.1038/s41598-021-90470-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 11/08/2022] Open
Abstract
Remote ischemic preconditioning (RIPC) involves deliberate, brief interruptions of blood flow to increase the tolerance of distant critical organs to ischemia. This study tests the effects of limb RIPC in a porcine model of controlled hemorrhage without replacement therapy simulating an extreme field situation of delayed evacuation to definitive care. Twenty-eight pigs (47 ± 6 kg) were assigned to: (1) control, no procedure (n = 7); (2) HS = hemorrhagic shock (n = 13); and (3) RIPC + HS = remote ischemic preconditioning followed by hemorrhage (n = 8). The animals were observed for 7 h after bleeding without fluid replacement. Survival rate between animals of the RIPC + HS group and those of the HS group were similar (HS, 6 of 13[46%]-vs-RIPC + HS, 4 of 8[50%], p = 0.86 by Chi-square). Animals of the RIPC + HS group had faster recovery of mean arterial pressure and developed higher heart rates without complications. They also had less decrease in pH and bicarbonate, and the increase in lactate began later. Global oxygen delivery was higher, and tissue oxygen extraction ratio lower, in RIPC + HS animals. These improvements after RIPC in hemodynamic and metabolic status provide essential substrates for improved cellular response after hemorrhage and reduction of the likelihood of potentially catastrophic consequences of the accompanying ischemia.
Collapse
|
61
|
Abstract
To characterize current evidence and current foci of perioperative clinical trials, we systematically reviewed Medline and identified perioperative trials involving 100 or more adult patients undergoing surgery and reporting renal end points that were published in high-impact journals since 2004. We categorized the 101 trials identified based on the nature of the intervention and summarized major trial findings from the five categories most applicable to perioperative management of patients. Trials that targeted ischemia suggested that increasing perioperative renal oxygen delivery with inotropes or blood transfusion does not reliably mitigate acute kidney injury (AKI), although goal-directed therapy with hemodynamic monitors appeared beneficial in some trials. Trials that have targeted inflammation or oxidative stress, including studies of nonsteroidal anti-inflammatory drugs, steroids, N-acetylcysteine, and sodium bicarbonate, have not shown renal benefits, and high-dose perioperative statin treatment increased AKI in some patient groups in two large trials. Balanced crystalloid intravenous fluids appear safer than saline, and crystalloids appear safer than colloids. Liberal compared with restrictive fluid administration reduced AKI in a recent large trial in open abdominal surgery. Remote ischemic preconditioning, although effective in several smaller trials, failed to reduce AKI in two larger trials. The translation of promising preclinical therapies to patients undergoing surgery remains poor, and most interventions that reduced perioperative AKI compared novel surgical management techniques or existing processes of care rather than novel pharmacologic interventions.
Collapse
Affiliation(s)
- David R McIlroy
- Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Marcos G Lopez
- Division of Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Frederic T Billings
- Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN; Division of Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
62
|
Krag AE, Blauenfeldt RA. Fibrinolysis and Remote Ischemic Conditioning: Mechanisms and Treatment Perspectives in Stroke. Semin Thromb Hemost 2021; 47:610-620. [PMID: 33878783 DOI: 10.1055/s-0041-1725095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Stroke is a leading cause of death and disability. Intravenous thrombolysis and mechanical thrombectomy have greatly improved outcomes in acute ischemic stroke (AIS). However, only a minority of patients receive reperfusion therapies, highlighting the need for novel neuroprotective therapies. Remote ischemic conditioning (RIC), consisting of brief, intermittent extremity occlusion and reperfusion induced with an inflatable cuff, is a potential neuroprotective therapy in acute stroke. The objective of this narrative review is to describe the effect of RIC on endogenous fibrinolysis and, from this perspective, investigate the potential of RIC in the prevention and treatment of stroke. A systematic literature search was performed in PubMed, and human studies in English were included. Seven studies had investigated the effect of RIC on fibrinolysis in humans. Long-term daily administration of RIC increased endogenous fibrinolysis, whereas a single RIC treatment did not acutely influence endogenous fibrinolysis. Fifteen studies had investigated the effect of RIC as a neuroprotective therapy in the prevention and treatment of stroke. Long-term RIC administration proved effective in reducing new cerebral vascular lesions in patients with established cerebrovascular disease. In patients with acute stroke, RIC was safe and feasible, though its clinical efficacy as a neuroprotectant is yet unproven. In conclusion, a single RIC treatment does not affect fibrinolysis in the acute phase, whereas long-term RIC administration may increase endogenous fibrinolysis. Increased endogenous fibrinolysis is unlikely to be the mediator of the acute neuroprotective effect of RIC in stroke patients, whereas it may partly explain the reduced stroke recurrence associated with long-term RIC treatment.
Collapse
Affiliation(s)
- Andreas Engel Krag
- Thrombosis and Hemostasis Research Unit, Aarhus University Hospital, Aarhus, Denmark
| | - Rolf Ankerlund Blauenfeldt
- Department of Neurology, Danish Stroke Center, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
63
|
Lassen TR, Hjortbak MV, Hauerslev M, Tonnesen PT, Kristiansen SB, Jensen RV, Bøtker HE. Influence of strain, age, origin, and anesthesia on the cardioprotective efficacy by local and remote ischemic conditioning in an ex vivo rat model. Physiol Rep 2021; 9:e14810. [PMID: 33818005 PMCID: PMC8020046 DOI: 10.14814/phy2.14810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background Local ischemic preconditioning (IPC) and remote ischemic conditioning (RIC) induced by brief periods of ischemia and reperfusion protect against ischemia‐reperfusion injury. Methods We studied the sensitivity to IR‐injury and the influence of strain, age, supplier, and anesthesia upon the efficacy of IPC and RIC in 7‐ and 16‐weeks‐old Sprague‐Dawley and Wistar rats from three different suppliers. The influence of sedation with a hypnorm and midazolam mixture (rodent mixture) and pentobarbiturate was compared. Results IPC attenuated infarct size in both 7‐weeks‐old Sprague–Dawley (48.4 ± 17.7% vs. 20.3 ± 6.9, p < 0.001) and 7‐weeks‐old Wistar (55.6 ± 10.9% vs. 26.8 ± 5.0%, p < 0.001) rats. Infarct size was larger in 16‐weeks‐old Sprague–Dawley rats, however, IPC still lowered infarct size (78.8 ± 9.2% vs. 58.3 ± 12.3%, p < 0.01). RIC reduced infarct sizes in 7‐weeks‐old Sprague–Dawley (75.3 ± 11.8% vs. 58.6 ± 8.9%, p < 0.05), but not in 7‐weeks‐old Wistar rats (31.7 ± 17.6% and 24.0 ± 12.6%, p = 0.2). In 16‐weeks‐old Sprague–Dawley rats, RIC did not induce protection (76.4 ± 5.5% and 73.2 ± 14.7%, p = 0.6). However, RIC induced protection in 16‐weeks‐old Wistar rats (45.2 ± 8.5% vs. 14.7 ± 10.8%, p < 0.001). RIC did not reduce infarct size in 7‐weeks‐old Sprague–Dawley rats from Charles River (62.0 ± 13.5% and 69.4 ± 10.4% p = 0.3) or 16‐weeks‐old Wistar rats from Janvier (50.7 ± 11.3 and 49.2 ± 16.2, p = 0.8). There was no difference between sedation with rodent mixture or pentobarbiturate. Conclusion The cardioprotective effect of IPC is consistent across rat strains independent of age, strain, and supplier. RIC seems to be less reproducible, but still yields protection across different rat strains. However, age, animal supplier, and anesthetics may modulate the sensitivity of IR‐injury and the response to RIC.
Collapse
Affiliation(s)
- Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Marie Vognstoft Hjortbak
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Marie Hauerslev
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Pernille Tilma Tonnesen
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | | | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
64
|
Erkens R, Totzeck M, Brum A, Duse D, Bøtker HE, Rassaf T, Kelm M. Endothelium-dependent remote signaling in ischemia and reperfusion: Alterations in the cardiometabolic continuum. Free Radic Biol Med 2021; 165:265-281. [PMID: 33497796 DOI: 10.1016/j.freeradbiomed.2021.01.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Intact endothelial function plays a fundamental role for the maintenance of cardiovascular (CV) health. The endothelium is also involved in remote signaling pathway-mediated protection against ischemia/reperfusion (I/R) injury. However, the transfer of these protective signals into clinical practice has been hampered by the complex metabolic alterations frequently observed in the cardiometabolic continuum, which affect redox balance and inflammatory pathways. Despite recent advances in determining the distinct roles of hyperglycemia, insulin resistance (InR), hyperinsulinemia, and ultimately diabetes mellitus (DM), which define the cardiometabolic continuum, our understanding of how these conditions modulate endothelial signaling remains challenging. It is widely accepted that endothelial cells (ECs) undergo functional changes within the cardiometabolic continuum. Beyond vascular tone and platelet-endothelium interaction, endothelial dysfunction may have profound negative effects on outcome during I/R. In this review, we summarize the current knowledge of the influence of hyperglycemia, InR, hyperinsulinemia, and DM on endothelial function and redox balance, their influence on remote protective signaling pathways, and their impact on potential therapeutic strategies to optimize protective heterocellular signaling.
Collapse
Affiliation(s)
- Ralf Erkens
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Amanda Brum
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Dragos Duse
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Hans Erik Bøtker
- Department of Cardiology, Institute of Clinical Medicine, Aarhus University Hospital, Denmark
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
65
|
Jarrard CP, Nagel MJ, Stray-Gundersen S, Tanaka H, Lalande S. Hypoxic preconditioning attenuates ischemia-reperfusion injury in young healthy adults. J Appl Physiol (1985) 2021; 130:846-852. [DOI: 10.1152/japplphysiol.00772.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ischemia-reperfusion injury induced by restoration of blood flow following occlusion impairs flow-mediated dilation, a marker of endothelium-dependent vasodilation. In young healthy adults, exposure to intermittent hypoxia, consisting of alternating short bouts of breathing hypoxic and normoxic air, before an ischemia-reperfusion injury significantly attenuated the reduction in flow-mediated dilation. Thus, hypoxic preconditioning represents a potential strategy to mitigate the effect of ischemia-reperfusion injury associated with ischemic events.
Collapse
Affiliation(s)
- Caitlin P. Jarrard
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Mercedes J. Nagel
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Sten Stray-Gundersen
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Hirofumi Tanaka
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Sophie Lalande
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
66
|
Baffour-Awuah B, Dieberg G, Pearson MJ, Smart NA. The effect of remote ischaemic conditioning on blood pressure response: A systematic review and meta-analysis. Int J Cardiol Hypertens 2021; 8:100081. [PMID: 33748739 PMCID: PMC7972960 DOI: 10.1016/j.ijchy.2021.100081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Previous work has evaluated the effect of remote ischaemic conditioning (RIC) in a number of clinical conditions (e.g. cardiac surgery and acute kidney injury), but only one analysis has examined blood pressure (BP) changes. While individual studies have reported the effects of acute bouts and repeated RIC exposure on resting BP, efficacy is equivocal. We conducted a systematic review and meta-analysis to evaluate the effects of acute and repeat RIC on BP. METHODS A systematic search was performed using PubMed, Web of Science, EMBASE, and Cochrane Library of Controlled Trials up until October 31, 2020. Additionally, manual searches of reference lists were performed. Studies that compared BP responses after exposing participants to either an acute bout or repeated cycles of RIC with a minimum one-week intervention period were considered. RESULTS Eighteen studies were included in this systematic review, ten examined acute effects while eight investigated repeat effects of RIC. Mean differences (MD) for outcome measures from acute RIC studies were: systolic BP 0.18 mmHg (95%CI -0.95, 1.31; p = 0.76), diastolic BP -0.43 mmHg (95%CI -2.36, 1.50; p = 0.66), MAP -1.73 mmHg (95%CI -3.11, -0.34; p = 0.01) and HR -1.15 bpm (95%CI -2.92, 0.62; p = 0.20). Only MAP was significantly reduced. Repeat RIC exposure showed non-significant change in systolic BP -3.23 mmHg (95%CI -6.57, 0.11; p = 0.06) and HR -0.16 bpm (95%CI -7.08, 6.77; p = 0.96) while diastolic BP -2.94 mmHg (95%CI -4.08, -1.79; p < 0.00001) and MAP -3.21 mmHg (95%CI -4.82, -1.61; p < 0.0001) were significantly reduced. CONCLUSIONS Our data suggests repeated, but not acute, RIC produced clinically meaningful reductions in diastolic BP and MAP.
Collapse
Affiliation(s)
- Biggie Baffour-Awuah
- Clinical Exercise Physiology, School of Science and Technology, Faculty of Science, Agriculture, Business and Law, University of New England, Armidale, NSW, 2351, Australia
| | - Gudrun Dieberg
- Biomedical Sciences, School of Science and Technology, Faculty of Science, Agriculture, Business and Law, University of New England, Armidale, NSW, 2351, Australia
| | - Melissa J. Pearson
- Clinical Exercise Physiology, School of Science and Technology, Faculty of Science, Agriculture, Business and Law, University of New England, Armidale, NSW, 2351, Australia
| | - Neil A. Smart
- Clinical Exercise Physiology, School of Science and Technology, Faculty of Science, Agriculture, Business and Law, University of New England, Armidale, NSW, 2351, Australia
| |
Collapse
|
67
|
Griffiths K, Lee JJ, Frenneaux MP, Feelisch M, Madhani M. Nitrite and myocardial ischaemia reperfusion injury. Where are we now? Pharmacol Ther 2021; 223:107819. [PMID: 33600852 DOI: 10.1016/j.pharmthera.2021.107819] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite major advances in technology and treatment, with coronary heart disease (CHD) being a key contributor. Following an acute myocardial infarction (AMI), it is imperative that blood flow is rapidly restored to the ischaemic myocardium. However, this restoration is associated with an increased risk of additional complications and further cardiomyocyte death, termed myocardial ischaemia reperfusion injury (IRI). Endogenously produced nitric oxide (NO) plays an important role in protecting the myocardium from IRI. It is well established that NO mediates many of its downstream functions through the 'canonical' NO-sGC-cGMP pathway, which is vital for cardiovascular homeostasis; however, this pathway can become impaired in the face of inadequate delivery of necessary substrates, in particular L-arginine, oxygen and reducing equivalents. Recently, it has been shown that during conditions of ischaemia an alternative pathway for NO generation exists, which has become known as the 'nitrate-nitrite-NO pathway'. This pathway has been reported to improve endothelial dysfunction, protect against myocardial IRI and attenuate infarct size in various experimental models. Furthermore, emerging evidence suggests that nitrite itself provides multi-faceted protection, in an NO-independent fashion, against a myriad of pathophysiologies attributed to IRI. In this review, we explore the existing pre-clinical and clinical evidence for the role of nitrate and nitrite in cardioprotection and discuss the lessons learnt from the clinical trials for nitrite as a perconditioning agent. We also discuss the potential future for nitrite as a pre-conditioning intervention in man.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jordan J Lee
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael P Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
68
|
Incognito AV, Millar PJ, Pyle WG. Remote ischemic conditioning for acute respiratory distress syndrome in COVID-19. Am J Physiol Lung Cell Mol Physiol 2021; 320:L331-L338. [PMID: 33404365 PMCID: PMC7938644 DOI: 10.1152/ajplung.00223.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome and subsequent respiratory failure remains the leading cause of death (>80%) in patients severely impacted by COVID-19. The lack of clinically effective therapies for COVID-19 calls for the consideration of novel adjunct therapeutic approaches. Though novel antiviral treatments and vaccination hold promise in control and prevention of early disease, it is noteworthy that in severe cases of COVID-19, addressing "run-away" inflammatory cascades are likely more relevant for improvement of clinical outcomes. Viral loads may decrease in severe, end-stage coronavirus cases, but a systemically damaging cytokine storm persists and mediates multiple organ injury. Remote ischemic conditioning (RIC) of the limbs has shown potential in recent years to protect the lungs and other organs against pathological conditions similar to that observed in COVID-19. We review the efficacy of RIC in protecting the lungs against acute injury and current points of consideration. The beneficial effects of RIC on lung injury along with other related cardiovascular complications are discussed, as are the limitations presented by sex and aging. This adjunct therapy is highly feasible, noninvasive, and proven to be safe in clinical conditions. If proven effective in clinical trials for acute respiratory distress syndrome and COVID-19, application in the clinical setting could be immediately implemented to improve outcomes.
Collapse
Affiliation(s)
- Anthony V Incognito
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Philip J Millar
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada.,Toronto General Research Institute, Toronto, Ontario, Canada
| | - W Glen Pyle
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.,IMPART Team Canada Investigator Network, Dalhousie Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| |
Collapse
|
69
|
Abstract
Perioperative cardioprotection aims to minimize the consequences of myocardial ischemia-reperfusion injury. In isolated tissue and animal experiments, several treatments have been identified providing cardioprotection. Some of these strategies have been confirmed in clinical proof-of-concept studies. However, the final translation of cardioprotective strategies to really improve clinical outcome has been disappointing: large randomized controlled clinical trials mostly revealed inconclusive, neutral, or negative results. This review provides an overview of the currently available evidence regarding clinical implications of perioperative cardioprotective therapies from an anesthesiological perspective, highlighting nonpharmacological as well as pharmacological strategies. We discuss reasons why translation of promising experimental results into clinical practice and outcome improvement is hampered by potential confounders and suggest future perspectives to overcome these limitations.
Collapse
|
70
|
Cho YJ, Nam K, Yoo SJ, Lee S, Bae J, Park JY, Kim HR, Kim TK, Jeon Y. Effects of remote ischemic preconditioning on platelet activation and reactivity in patients undergoing cardiac surgery using cardiopulmonary bypass: a randomized controlled trial. Platelets 2020; 33:123-131. [PMID: 33307907 DOI: 10.1080/09537104.2020.1856362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
During cardiopulmonary bypass (CPB), platelet activation and dysfunction are associated with adverse outcomes. Remote ischemic preconditioning (RIPC) has been shown to attenuate platelet activation. We evaluated the effects of RIPC on platelet activation during CPB in patients undergoing cardiac surgery. Among 58 randomized patients, 26 in the RIPC group and 28 in the sham-RIPC group were analyzed. RIPC consisted of 4 cycles of 5-min ischemia induced by inflation of pneumatic cuff pressure to 200 mmHg, followed by 5-min reperfusion comprising deflation of the cuff on the upper arm. Platelet activation was assessed using flow cytometry analysis of platelet activation markers. The primary endpoint was the AUC of CD62P expression during the first 3 h after initiation of CPB. Secondary outcomes were the AUC of PAC-1 expression and monocyte-platelet aggregates (MPA) during 3 h of CPB. The AUCs of CD62P expression during 3 h after initiation of CPB were 219.4 ± 43.9 and 211.0 ± 41.2 MFI in the RIPC and sham-RIPC groups, respectively (mean difference, 8.42; 95% CI, -14.8 and 31.7 MFI; p =.471). The AUCs of PAC-1 expression and MPA did not differ between groups. RIPC did not alter platelet activation and reactivity during CPB in patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Youn Joung Cho
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Karam Nam
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sol Ji Yoo
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seohee Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jinyoung Bae
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Young Park
- FACS Core Facility, Seoul National University College of Medicine, Seoul, Korea
| | - Hang-Rae Kim
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Kyong Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Yunseok Jeon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
71
|
Hyngstrom AS, Nguyen JN, Wright MT, Tarima SS, Schmit BD, Gutterman DD, Durand MJ. Two weeks of remote ischemic conditioning improves brachial artery flow mediated dilation in chronic stroke survivors. J Appl Physiol (1985) 2020; 129:1348-1354. [PMID: 33090908 DOI: 10.1152/japplphysiol.00398.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Many stroke survivors have reduced cardiorespiratory fitness as a result of their stroke. Ischemic conditioning (IC) is a noninvasive, cost-effective, easy-to-administer intervention that can be performed at home and has been shown to improve both motor function in stroke survivors and vascular endothelial function in healthy individuals. In this study, we examined the effects of 2 wk of remote IC (RIC) on brachial artery flow mediated dilation (FMD) in chronic stroke survivors. We hypothesized that FMD would be improved following RIC compared with a sham RIC control group. This was a prospective, randomized, double-blinded, controlled study. Twenty-four chronic stroke survivors (>6 mo after stroke) were enrolled and randomized to receive either RIC or sham RIC on their affected thigh every other day for 2 wk. For the RIC group, a blood pressure cuff was inflated to 225 mmHg for 5 min, followed by 5 min of recovery, and repeated a total of five times per session. For the sham RIC group, the inflation pressure was 10 mmHg. Brachial artery FMD was assessed on the nonaffected arm at study enrollment and following the 2-wk intervention period. Nine men and fourteen women completed all study procedures. Brachial artery FMD increased from 5.4 ± 4.8 to 7.8 ± 4.4% (P = 0.030; n = 12) in the RIC group, while no significant change was observed in the sham RIC group (3.5 ± 3.9% pretreatment versus 2.4 ± 3.1% posttreatment; P = 0.281, n = 11). Two weeks of RIC increases brachial artery FMD in chronic stroke survivors.NEW & NOTEWORTHY In this study, we report that 2 wk of remote ischemic conditioning (RIC) improves brachial artery flow-mediated dilation in chronic stroke survivors. Because poor cardiovascular health puts stroke survivors at a heightened risk for recurrent stroke and other cardiovascular events, an intervention that is simple, cost-effective, and easy to perform like RIC holds promise as a means to improve cardiovascular health in this at-risk population.
Collapse
Affiliation(s)
| | - Jennifer N Nguyen
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael T Wright
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sergey S Tarima
- Institute of Health and Equity, Division of Biostatistics, Medical College of Wisconsin Milwaukee, Wisconsin
| | - Brian D Schmit
- Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David D Gutterman
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Matthew J Durand
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
72
|
Kleinbongard P, Bøtker HE, Ovize M, Hausenloy DJ, Heusch G. Co-morbidities and co-medications as confounders of cardioprotection-Does it matter in the clinical setting? Br J Pharmacol 2020; 177:5252-5269. [PMID: 31430831 PMCID: PMC7680006 DOI: 10.1111/bph.14839] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/26/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023] Open
Abstract
The translation of cardioprotection from robust experimental evidence to beneficial clinical outcome for patients suffering acute myocardial infarction or undergoing cardiovascular surgery has been largely disappointing. The present review attempts to critically analyse the evidence for confounders of cardioprotection in patients with acute myocardial infarction and in patients undergoing cardiovascular surgery. One reason that has been proposed to be responsible for such lack of translation is the confounding of cardioprotection by co-morbidities and co-medications. Whereas there is solid experimental evidence for such confounding of cardioprotection by single co-morbidities and co-medications, the clinical evidence from retrospective analyses of the limited number of clinical data is less robust. The best evidence for interference of co-medications is that for platelet inhibitors to recruit cardioprotection per se and thus limit the potential for further protection from myocardial infarction and for propofol anaesthesia to negate the protection from remote ischaemic conditioning in cardiovascular surgery. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular CenterUniversity of Essen Medical SchoolEssenGermany
| | - Hans Erik Bøtker
- Department of CardiologyAarhus University Hospital SkejbyAarhusDenmark
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Hospices Civils de LyonLyonFrance
| | - Derek J. Hausenloy
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingapore
- National Heart Research Institute SingaporeNational Heart CentreSingapore
- Yong Loo Lin School of MedicineNational University SingaporeSingapore
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Research and DevelopmentThe National Institute of Health Research University College London Hospitals Biomedical Research CentreLondonUK
- Tecnologico de MonterreyCentro de Biotecnologia‐FEMSAMonterreyNuevo LeonMexico
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular CenterUniversity of Essen Medical SchoolEssenGermany
| |
Collapse
|
73
|
Uutela A, Helanterä I, Lemström K, Passov A, Syrjälä S, Åberg F, Mäkisalo H, Nordin A, Lempinen M, Sallinen V. Randomised sham-controlled double-blind trial evaluating remote ischaemic preconditioning in solid organ transplantation: a study protocol for the RIPTRANS trial. BMJ Open 2020; 10:e038340. [PMID: 33199419 PMCID: PMC7670950 DOI: 10.1136/bmjopen-2020-038340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Remote ischaemic preconditioning (RIPC) using a non-invasive pneumatic tourniquet is a potential method for reducing ischaemia-reperfusion injury. RIPC has been extensively studied in animal models and cardiac surgery, but scarcely in solid organ transplantation. RIPC could be an inexpensive and simple method to improve function of transplanted organs. Accordingly, we aim to study whether RIPC performed in brain-dead organ donors improves function and longevity of transplanted organs. METHODS AND ANALYSES RIPTRANS is a multicentre, sham-controlled, parallel group, randomised superiority trial comparing RIPC intervention versus sham-intervention in brain-dead organ donors scheduled to donate at least one kidney. Recipients of the organs (kidney, liver, pancreas, heart, lungs) from a randomised donor will be included provided that they give written informed consent. The RIPC intervention is performed by inflating a thigh tourniquet to 300 mm Hg 4 times for 5 min. The intervention is done two times: first right after the declaration of brain death and second immediately before transferring the donor to the operating theatre. The sham group receives the tourniquet, but it is not inflated. The primary endpoint is delayed graft function (DGF) in kidney allografts. Secondary endpoints include short-term functional outcomes of transplanted organs, rejections and graft survival in various time points up to 20 years. We aim to show that RIPC reduces the incidence of DGF from 25% to 15%. According to this, the sample size is set to 500 kidney transplant recipients. ETHICS AND DISSEMINATION This study has been approved by Helsinki University Hospital Ethics Committee and Helsinki University Hospital's Institutional Review Board. The study protocol was be presented at the European Society of Organ Transplantation congress in Copenhagen 14-15 September 2019. The study results will be submitted to an international peer-reviewed scientific journal for publication. TRIAL REGISTRATION NUMBER NCT03855722.
Collapse
Affiliation(s)
- Aki Uutela
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ilkka Helanterä
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Karl Lemström
- Department of Cardiothoracic Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Arie Passov
- Department of Perioperative, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Simo Syrjälä
- Department of Cardiothoracic Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Fredrik Åberg
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heikki Mäkisalo
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Arno Nordin
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Marko Lempinen
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ville Sallinen
- Department of Transplantation and Liver Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
74
|
Trachte TE, Hemenway BA, Van Guilder GP. Reduced effect of ischemic preconditioning against endothelial ischemia-reperfusion injury with cardiovascular risk factors in humans. J Hum Hypertens 2020; 35:870-879. [PMID: 33168942 DOI: 10.1038/s41371-020-00440-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/23/2020] [Accepted: 10/26/2020] [Indexed: 11/09/2022]
Abstract
The extent that clustered CVD risk factors interfere with ischemic preconditioning (IPC) to protect against microvascular endothelial dysfunction with ischemia-reperfusion (I/R) injury in humans is unclear. We hypothesized that adults with a clustered burden of ≥3 CVD risk factors would demonstrate a reduced capacity of IPC to protect endothelial function with I/R injury. Twenty-two (age: 45 ± 14 year) adults [12 healthy controls; 10 raised risk (10-year FRS risk score ~3%)] were studied using a 2 × 2 randomized cross-over design. Pulse arterial tonometry was used to assess microvascular endothelium-dependent vasodilation during reactive hyperemia in response to endothelial I/R injury (20 min brachial artery occlusion/45 min reperfusion) that was preceded by remote IPC (3 × 5 min ischemia/reperfusion) or mock IPC. In both groups, microvascular reactive hyperemia was reduced ~20% (both P < 0.01) after endothelial I/R injury without remote IPC. However, in control subjects remote IPC prevented endothelial I/R injury (from baseline reactive hyperemic ratio: 2.1 ± 0.4 AU to post I/R injury: 2.5 ± 0.5 AU; P = 0.09). In contrast, the reactive hyperemia ratio in raised risk subjects was significantly reduced from 2.2 ± 0.6 AU to 1.9 ± 0.5 AU (P = 0.0087) despite attempts to induce protection by remote IPC, with the magnitude of reduction similar to their mock IPC trial. The magnitude of remote IPC-mediated endothelial protection against I/R injury was inversely related to the number of risk factors. CVD risk factors diminish the effect of IPC to protect the microvasculature from I/R injury in humans. Translating IPC to clinical practice for vasculoprotection will continue to be challenging in patients with increased CVD risk.
Collapse
Affiliation(s)
- Tiffany E Trachte
- Exercise & Sport Science Department, Western Colorado University, Paul Wright Gym 209, 1 Western Way, Gunnison, CO, 81231, USA
| | - Brian A Hemenway
- Exercise & Sport Science Department, Western Colorado University, Paul Wright Gym 209, 1 Western Way, Gunnison, CO, 81231, USA
| | - Gary P Van Guilder
- Exercise & Sport Science Department, Western Colorado University, Paul Wright Gym 209, 1 Western Way, Gunnison, CO, 81231, USA.
| |
Collapse
|
75
|
Bashir SO, Morsy MD, El Agamy DF. Two episodes of remote ischemia preconditioning improve motor and sensory function of hind limbs after spinal cord ischemic injury. J Spinal Cord Med 2020; 43:878-887. [PMID: 30985269 PMCID: PMC7801032 DOI: 10.1080/10790268.2019.1600829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Objectives: To investigate the effect of one and two remote ischemia preconditioning episodes (1-RIPC or 2-RIPC, respectively) on neuro-protection after spinal cord ischemic injury (SCI) in rats. Design: Experimental animal study. Setting: College of Medicine, King Khalid University, Abha, KSA. Interventions: Male rats (n = 10/group) were divided into control, sham, SCIRI, 1-RIPC + SCIRI, and 2-RIPC + SCIRI. SCI was induced by aortic ligation for 45 min and each RIPC episode was induced by 3 cycles of 10 min ischemia/10 min perfusion. The two preconditioning procedures were separated by 24 h. Outcome measures: after 48 h of RIPC procedure, Tarlov's test, withdrawal from the painful stimulus and placing/stepping reflex (SPR) were used to evaluate the hind limbs neurological function. SC homogenates were used to measure various biochemical parameters. Results: Motor and sensory function of hind limbs were significantly improved and levels of MDA, AOPPs, PGE2, TNF-α, and IL-6, as well as the activity of SOD, was significantly decreased in SC tissue in either 1 or 2 episodes of RIPC intervention. Concomitantly, levels of total nitrate/nitrite and eNOS activity were significantly increased in both groups. Interestingly, except for activity of SOD, eNOS and levels of nitrate/nitrite, the improvements in all neurological biochemical endpoint were more profound in 2-RIPC + SCIRI compared with 1-RIPC + SCIRI. Conclusion: applying two preconditioning episodes of 3 cycles of 10 min ischemia/10 min perfusion, separated by 24 h, boost the neuro-protection effect of RIPC maneuver in rats after ischemic induced SCI in rats.
Collapse
Affiliation(s)
- Salah Omar Bashir
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Darwesh Morsy
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia,Department of Physiology, College of Medicine, Menoufia University, Shebeen Alkoom, Egypt,Correspondence to: Mohamed Darwesh Morsy, Department of Physiology, College of Medicine, King Khalid University, Abha61421, Saudi Arabia; Mobile Number: +966544495223; Fax: +966+966172251690; E-mail:
| | - Dalia Fathy El Agamy
- Department of Physiology, College of Medicine, Menoufia University, Shebeen Alkoom, Egypt
| |
Collapse
|
76
|
Raval RN, Small O, Magsino K, Chakravarthy V, Austin B, Applegate R, Dorotta I. Remote Ischemic Pre-conditioning in Subarachnoid Hemorrhage: A Prospective Pilot Trial. Neurocrit Care 2020; 34:968-973. [PMID: 33051793 DOI: 10.1007/s12028-020-01122-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/21/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cerebral injury from aneurysmal subarachnoid hemorrhage (aSAH) is twofold. The initial hemorrhage causes much of the injury; secondary injury can occur from delayed cerebral ischemia (DCI). Remote ischemic preconditioning (RIPC) is a mechanism of organ protection in response to transient ischemia within a distant organ. This pilot trial sought to apply RIPC in patients with aSAH to evaluate its effect on secondary cerebral injury and resultant outcomes. METHODS Patients were randomized to the high-pressure occlusion group (HPO) or the low-pressure occlusion group (LPO). Lower extremity RIPC treatment was initiated within 72 h of symptom onset and every other day for 14 days or until Intensive Care Unit (ICU) discharge. In HPO, each treatment consisted of 4 five-minute cycles of manual blood pressure cuff inflation with loss of distal pulses. LPO received cuff inflation with lower pressures while preserving distal pulses. Retrospectively matched controls were also analyzed. Efficacy of treatment was measured by total days spent in vasospasm out of study enrollment days, hospital and ICU length of stay (LOS), cerebral infarction, one and six month modified Rankin score, and mortality. RESULTS The final analysis included 33 patients with 11 in each group. Patient demographics, aneurysm location, admission airway status, Glasgow Coma Scale (GCS), modified Rankin score, Hunt and Hess score, modified Fisher Score and aneurysm management were not significantly different between groups. Hospital and ICU LOS was shorter in LPO compared to the control (p = 0·0468 and p = 0·0409, respectively). Total vasospasm days/study enrollment days, cerebral infarction, one and six month modified Rankin score, and mortality were not significantly different between the groups. CONCLUSIONS This pilot trial did demonstrate feasibility and safety. The shortened LOS in the LPO may implicate a protective role of RIPC and warrants future study.
Collapse
Affiliation(s)
- Ronak N Raval
- Department of Anesthesiology and Critical Care Center, Loma Linda University Medical Center, 11234 Anderson Street, Loma Linda, CA, 92354, USA. .,Department of Surgery, VA Loma Linda Healthcare System, Loma Linda, USA.
| | - Oliver Small
- Department of Anesthesiology, Swedish Medical Center, Seattle, USA
| | - Kristel Magsino
- Department of Anesthesiology and Critical Care Center, Loma Linda University Medical Center, 11234 Anderson Street, Loma Linda, CA, 92354, USA
| | - Vikram Chakravarthy
- Department of Neurosurgery, Cleveland Clinic of Case Western Reserve School of Medicine, Cleveland, USA
| | - Briahnna Austin
- Department of Anesthesiology and Critical Care Center, Loma Linda University Medical Center, 11234 Anderson Street, Loma Linda, CA, 92354, USA
| | - Richard Applegate
- Department of Anesthesiology, Davis Medical Center, University of California, Sacramento, USA
| | - Ihab Dorotta
- Department of Anesthesiology and Critical Care Center, Loma Linda University Medical Center, 11234 Anderson Street, Loma Linda, CA, 92354, USA
| |
Collapse
|
77
|
Drury NE, Bi R, Woolley RL, Stickley J, Morris KP, Montgomerie J, van Doorn C, Dunn WB, Madhani M, Ives NJ, Kirchhof P, Jones TJ. Bilateral Remote Ischaemic Conditioning in Children (BRICC) trial: protocol for a two-centre, double-blind, randomised controlled trial in young children undergoing cardiac surgery. BMJ Open 2020; 10:e042176. [PMID: 33033035 PMCID: PMC7542918 DOI: 10.1136/bmjopen-2020-042176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Myocardial protection against ischaemic-reperfusion injury is a key determinant of heart function and outcome following cardiac surgery in children. However, with current strategies, myocardial injury occurs routinely following aortic cross-clamping, as demonstrated by the ubiquitous rise in circulating troponin. Remote ischaemic preconditioning, the application of brief, non-lethal cycles of ischaemia and reperfusion to a distant organ or tissue, is a simple, low-risk and readily available technique which may improve myocardial protection. The Bilateral Remote Ischaemic Conditioning in Children (BRICC) trial will assess whether remote ischaemic preconditioning, applied to both lower limbs immediately prior to surgery, reduces myocardial injury in cyanotic and acyanotic young children. METHODS AND ANALYSIS The BRICC trial is a two-centre, double-blind, randomised controlled trial recruiting up to 120 young children (age 3 months to 3 years) undergoing primary repair of tetralogy of Fallot or surgical closure of an isolated ventricular septal defect. Participants will be randomised in a 1:1 ratio to either bilateral remote ischaemic preconditioning (3×5 min cycles) or sham immediately prior to surgery, with follow-up until discharge from hospital or 30 days, whichever is sooner. The primary outcome is reduction in area under the time-concentration curve for high-sensitivity (hs) troponin-T release in the first 24 hours after aortic cross-clamp release. Secondary outcome measures include peak hs-troponin-T, vasoactive inotrope score, arterial lactate and central venous oxygen saturations in the first 12 hours, and lengths of stay in the paediatric intensive care unit and the hospital. ETHICS AND DISSEMINATION The trial was approved by the West Midlands-Solihull National Health Service Research Ethics Committee (16/WM/0309) on 5 August 2016. Findings will be disseminated to the academic community through peer-reviewed publications and presentation at national and international meetings. Parents will be informed of the results through a newsletter in conjunction with a local charity. TRIAL REGISTRATION NUMBER ISRCTN12923441.
Collapse
Affiliation(s)
- Nigel E Drury
- Paediatric Cardiac Surgery, Birmingham Children's Hospital, Birmingham, West Midlands, UK
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Rehana Bi
- Paediatric Cardiac Surgery, Birmingham Children's Hospital, Birmingham, West Midlands, UK
- Paediatric Intensive Care, Birmingham Children's Hospital, Birmingham, West Midlands, UK
| | - Rebecca L Woolley
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, West Midlands, UK
- Institute of Applied Health Research, University of Birmingham, Birmingham, West Midlands, UK
| | - John Stickley
- Paediatric Cardiac Surgery, Birmingham Children's Hospital, Birmingham, West Midlands, UK
| | - Kevin P Morris
- Paediatric Intensive Care, Birmingham Children's Hospital, Birmingham, West Midlands, UK
- Institute of Applied Health Research, University of Birmingham, Birmingham, West Midlands, UK
| | - James Montgomerie
- Paediatric Cardiac Anaesthesia, Birmingham Children's Hospital, Birmingham, West Midlands, UK
| | - Carin van Doorn
- Congenital Cardiac Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, West Yorkshire, UK
| | - Warwick B Dunn
- School of Biosciences, University of Birmingham, Birmingham, West Midlands, UK
- Phenome Centre Birmingham, University of Birmingham, Birmingham, West Midlands, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Natalie J Ives
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, West Midlands, UK
- Institute of Applied Health Research, University of Birmingham, Birmingham, West Midlands, UK
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
- Cardiology, University Heart and Vascular Center, UKE, Hamburg, Germany
| | - Timothy J Jones
- Paediatric Cardiac Surgery, Birmingham Children's Hospital, Birmingham, West Midlands, UK
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
78
|
Koike Y, Li B, Ganji N, Zhu H, Miyake H, Chen Y, Lee C, Janssen Lok M, Zozaya C, Lau E, Lee D, Chusilp S, Zhang Z, Yamoto M, Wu RY, Inoue M, Uchida K, Kusunoki M, Delgado-Olguin P, Mertens L, Daneman A, Eaton S, Sherman PM, Pierro A. Remote ischemic conditioning counteracts the intestinal damage of necrotizing enterocolitis by improving intestinal microcirculation. Nat Commun 2020; 11:4950. [PMID: 33009377 PMCID: PMC7532542 DOI: 10.1038/s41467-020-18750-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease of premature infants with high mortality rate, indicating the need for precision treatment. NEC is characterized by intestinal inflammation and ischemia, as well derangements in intestinal microcirculation. Remote ischemic conditioning (RIC) has emerged as a promising tool in protecting distant organs against ischemia-induced damage. However, the effectiveness of RIC against NEC is unknown. To address this gap, we aimed to determine the efficacy and mechanism of action of RIC in experimental NEC. NEC was induced in mouse pups between postnatal day (P) 5 and 9. RIC was applied through intermittent occlusion of hind limb blood flow. RIC, when administered in the early stages of disease progression, decreases intestinal injury and prolongs survival. The mechanism of action of RIC involves increasing intestinal perfusion through vasodilation mediated by nitric oxide and hydrogen sulfide. RIC is a viable and non-invasive treatment strategy for NEC. Necrotizing enterocolitis (NEC) is one of the most lethal gastrointestinal emergencies in neonates needing precision treatment. Here the authors show that remote ischemic conditioning is a non-invasive therapeutic method that enhances blood flow in the intestine, reduces damage, and improves NEC outcome.
Collapse
Affiliation(s)
- Yuhki Koike
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Bo Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Niloofar Ganji
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Haitao Zhu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hiromu Miyake
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yong Chen
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carol Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Maarten Janssen Lok
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carlos Zozaya
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ethan Lau
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dorothy Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sinobol Chusilp
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zhen Zhang
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Masaya Yamoto
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Richard Y Wu
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mikihiro Inoue
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Keiichi Uchida
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masato Kusunoki
- Departments of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Paul Delgado-Olguin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
| | - Luc Mertens
- The Labatt Family Heart Center, Cardiology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Alan Daneman
- Department of Diagnostic Imaging, Division of Nuclear Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Philip M Sherman
- Cell Biology Program, Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON, Canada.,Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Agostino Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Division of General and Thoracic Surgery, Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
79
|
Caiazzo G, Musci RL, Frediani L, Umińska J, Wanha W, Filipiak KJ, Kubica J, Navarese EP. State of the Art: No-Reflow Phenomenon. Cardiol Clin 2020; 38:563-573. [PMID: 33036718 DOI: 10.1016/j.ccl.2020.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Primary percutaneous coronary intervention is the preferred reperfusion strategy for the management of acute ST-segment elevation myocardial infarction. No reflow is characterized by the inadequate myocardial perfusion of a given segment without angiographic evidence of persistent mechanical obstruction of epicardial vessels. Both pharmacologic and device-based strategies have been tested to resolve coronary no reflow. This article provides an updated overview of the no-reflow phenomenon, discussing clinical evidence and ongoing investigations of existing and novel therapeutic strategies to counteract it.
Collapse
Affiliation(s)
- Gianluca Caiazzo
- ICCU, San Giuseppe Moscati Hospital, ASL CE, Via Gramsci 1, Aversa 81031, Italy
| | - Rita Leonarda Musci
- Department of Cardiology, Azienda Ospedaliera Bonomo, Viale Istria, Andria BT 76123, Italy
| | - Lara Frediani
- Department of Cardiology, Livorno Hospital, Azienda Usl Toscana Nord-Ovest, Ospedali Riuniti di Livorno, Viale Vittorio Alfieri, 36, Livorno LI 57124, Italy
| | - Julia Umińska
- Department of Cardiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, SIRIO MEDICINE Network, ul. Jagiellońska 13-15, Bydgoszcz 85-067, Poland
| | - Wojciech Wanha
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, ul. Józefa Poniatowskiego 15, Kato 40-055, Katowice, Poland
| | - Krzysztof J Filipiak
- Department of Cardiology, Medical University of Warsaw, Żwirki i Wigury 61, Warszawa 02-091, Poland
| | - Jacek Kubica
- Department of Cardiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, SIRIO MEDICINE Network, ul. Jagiellońska 13-15, Bydgoszcz 85-067, Poland
| | - Eliano Pio Navarese
- Department of Cardiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, SIRIO MEDICINE Network, ul. Jagiellońska 13-15, Bydgoszcz 85-067, Poland; University of Alberta, 116 Street & 85 Avenue, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
80
|
Abstract
Preconditioning, a milestone concept in the cardiovascular sciences introduced 32 years back by Murry. This concept opened a new era in the field of organ protection. To start with extensive studies done on ischemic preconditioning for myocardial protection, ischemic preconditioning is an endogenous science of cellular kinetics. Several components in signal transduction cascade have been identified but still some mechanisms not yet revealed. Anesthetic preconditioning also contributed a lot for myocardial protection and concreted the concept of preconditioning. We, with an inquisitive brain meticulously persuing newer methods of cardioprotection. Remote ischemic preconditioning (RIPC) is a brilliant example of it. RIPC can be future of cardioprotection, clinical trials and studies proved the benefits but yet to conclude the superiority of RIPC over myocardial ischemic cardioprotection. This review is an attempt to reveal this extraordinary concept with its basic cellular kinetics, methods, and recent trends.
Collapse
Affiliation(s)
| | - Suhrid R Annachhatre
- Department of CVTS, MCRI MGM Medical College and Hospital, Aurangabad, Maharashtra, India
| |
Collapse
|
81
|
Giblett JP, Bulluck H. Cardioprotection for Acute MI in Light of the CONDI2/ERIC-PPCI Trial: New Targets Needed. ACTA ACUST UNITED AC 2020; 15:e13. [PMID: 32944081 PMCID: PMC7479528 DOI: 10.15420/icr.2020.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023]
Abstract
Protection against ischaemia-reperfusion injury after revascularisation in acute myocardial infarction remains an enigma. Many targets have been identified, but after the failure of the recent Effect of Remote Ischaemic Conditioning on Clinical Outcomes in ST-elevation Myocardial Infarction Patients Undergoing Primary Percutaneous Coronary Intervention (CONDI2/ERIC-PPCI) trial to show translation to clinical benefit, there is still no pharmacological or mechanical strategy that has translated to clinical practice. This article addresses the results of the CONDI2/ERIC-PPCI trial in the context of previous studies of ischaemic conditioning, and then considers the prospects for other potential targets of cardioprotection. Finally, the authors examine the pitfalls and challenges in trial design for future investigation of cardioprotective strategies. In particular, this article highlights the need for careful endpoint and patient selection, as well as the need to pay attention to the biology of cardioprotection during the study.
Collapse
Affiliation(s)
- Joel P Giblett
- Department of Cardiology, Liverpool Heart and Chest Hospital Liverpool, UK
| | | |
Collapse
|
82
|
Choi EK, Jung H, Jeon S, Lim JA, Lee J, Kim H, Hong SW, Jang MH, Lim DG, Kwak KH. Role of Remote Ischemic Preconditioning in Hepatic Ischemic Reperfusion Injury. Dose Response 2020; 18:1559325820946923. [PMID: 32848526 PMCID: PMC7427033 DOI: 10.1177/1559325820946923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/25/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
The effect of remote ischemic preconditioning (RIPC) has been proposed that mediates the protective response in ischemia reperfusion injury (IRI) of various organs. In this study, we investigated the effect of RIPC in hepatic IRI, by assessing biomarker of oxidative stress and inflammatory cytokines. Moreover, we intended to demonstrate any such protective effect through nitric oxide (NO). Twenty-five rats were divided into the 5 groups: (1) Sham; (2) RIPC; (3) hepatic IRI; (4) RIPC + hepatic IRI; (5) C-PTIO, 2-(4-carboxyphenyl)-4,5dihydro-4,4,5,5-tetramethyl-1H-imidazolyl-1-oxy-3oxide, + RIPC + hepatic IRI. RIPC downregulated the level of aspartate aminotransferase (AST), alanine aminotransferase (ALT), histologic damage, and activity of Malondialdehyde (MDA). However, there was no significant reduction in the level of tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa B (NF-κB). AST and ALT levels, and hepatic tissue morphology in the C-PTIO group showed a significant improvement compared to those of the RIPC + hepatic IRI group. The application of RIPC before hepatic ischemia downregulated the oxidative stress, not the inflammatory cytokines. Moreover, these protective effect of RIPC would be mediated through the activation of NO as well as anti-oxidant effect.
Collapse
Affiliation(s)
- Eun Kyung Choi
- Department of Anesthesiology and Pain Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Hoon Jung
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sungmin Jeon
- Department of Anesthesiology and Pain Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jung A Lim
- Department of Anesthesiology and Pain Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jungwon Lee
- Department of Anesthesiology and Pain Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Hyunjee Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seong Wook Hong
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Min Hye Jang
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Dong Gun Lim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung Hwa Kwak
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
83
|
Rytter N, Carter H, Piil P, Sørensen H, Ehlers T, Holmegaard F, Tuxen C, Jones H, Thijssen D, Gliemann L, Hellsten Y. Ischemic Preconditioning Improves Microvascular Endothelial Function in Remote Vasculature by Enhanced Prostacyclin Production. J Am Heart Assoc 2020; 9:e016017. [PMID: 32750305 PMCID: PMC7792245 DOI: 10.1161/jaha.120.016017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The mechanisms underlying the effect of preconditioning on remote microvasculature remains undisclosed. The primary objective was to document the remote effect of ischemic preconditioning on microvascular function in humans. The secondary objective was to test if exercise also induces remote microvascular effects. METHODS AND RESULTS A total of 12 healthy young men and women participated in 2 experimental days in a random counterbalanced order. On one day the participants underwent 4×5 minutes of forearm ischemic preconditioning, and on the other day they completed 4×5 minutes of hand-grip exercise. On both days, catheters were placed in the brachial and femoral artery and vein for infusion of acetylcholine, sodium nitroprusside, and epoprostenol. Vascular conductance was calculated from blood flow measurements with ultrasound Doppler and arterial and venous blood pressures. Ischemic preconditioning enhanced (P<0.05) the remote vasodilator response to intra-arterial acetylcholine in the leg at 5 and 90 minutes after application. The enhanced response was associated with a 6-fold increase (P<0.05) in femoral venous plasma prostacyclin levels and with a transient increase (P<0.05) in arterial plasma levels of brain-derived neurotrophic factor and vascular endothelial growth factor. In contrast, hand-grip exercise did not influence remote microvascular function. CONCLUSIONS These findings demonstrate that ischemic preconditioning of the forearm improves remote microvascular endothelial function and suggest that one of the underlying mechanisms is a humoral-mediated potentiation of prostacyclin formation.
Collapse
Affiliation(s)
- Nicolai Rytter
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Howard Carter
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Peter Piil
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Henrik Sørensen
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark.,Department of Anesthesia Centre for Cancer and Organ Diseases Rigshospitalet Copenhagen Denmark
| | - Thomas Ehlers
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Frederik Holmegaard
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Christoffer Tuxen
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Helen Jones
- Research Institute for Sport and Exercise Sciences Liverpool John Moores University Liverpool United Kingdom
| | - Dick Thijssen
- Research Institute for Sport and Exercise Sciences Liverpool John Moores University Liverpool United Kingdom.,Department of Physiology Radboud Institute for Health Sciences Nijmegen The Netherlands
| | - Lasse Gliemann
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| | - Ylva Hellsten
- Section of Integrative Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Denmark
| |
Collapse
|
84
|
Gardner RN, Sabino-Carvalho JL, Kim J, Vianna LC, Lang JA. Two weeks of remote ischaemic preconditioning alters sympathovagal balance in healthy humans. Exp Physiol 2020; 105:1500-1506. [PMID: 32691505 DOI: 10.1113/ep088789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the central question of this study? Delayed cardiovascular responses occur following a single bout of remote ischaemic preconditioning (RIPC). Is heart rate variability (HRV), a surrogate marker of cardiac vagal control, able to detect a delayed effect after a single bout of RIPC? Do repeated bouts of RIPC further alter HRV? What is the main finding and its importance? Indices of HRV indicated a shift in sympathovagal balance toward greater parasympathetic activity following 2 weeks of RIPC but not after a single bout of RIPC. Thus, repeated bouts of RIPC were necessary to elicit changes in autonomic function. ABSTRACT Remote ischaemic preconditioning (RIPC), induced by brief periods of ischaemia followed by reperfusion, protects against ischaemia-reperfusion injury and improves microvascular function. However, the effect of RIPC on autonomic function remains unclear. We hypothesized that RIPC, administered as a single bout or repeated over a 2-week period, will increase markers of cardiac vagal control measured by heart rate variability (HRV). Thirty-two young adults performed a single bout (n = 13), repeated bouts (n = 11), or served as a time control (n = 8). RIPC sessions consisted of four repetitions of 5 min unilateral brachial artery occlusion interspersed by 5 min of reperfusion. For the single bout protocol, resting lead II electrocardiogram (ECG) was collected before and 24, 48, 72 and 168 h post-RIPC. The repeated bout protocol consisted of three 4-day periods of RIPC training, each interspersed by a 1-day break. Similar to time controls, ECG was collected before and 24 h after the last RIPC bout. HRV was analysed by power spectral density and symbolic dynamics using 350-beat ECG segments. After a single bout of RIPC, no changes in HRV were observed at any time point (P > 0.05). After 2 weeks of repeated RIPC, the percentage of zero-variation fragments (baseline = 13.1 ± 1.9%, post-RIPC = 6.9 ± 1.5%, P < 0.05) and the LF/HF ratio decreased (baseline = 1.1 ± 0.2, post-RIPC = 0.7 ± 0.1, P < 0.01), whereas the percentage of two-variation fragments increased (baseline = 42.9 ± 3.6%, post-RIPC = 52.5 ± 3.0%, P < 0.01). These data indicate that repeated RIPC is necessary to elicit changes in sympathovagal balance, specifically resulting in increased vagal and decreased sympathetic activity.
Collapse
Affiliation(s)
| | - Jeann L Sabino-Carvalho
- Department of Kinesiology, Iowa State University, Ames, IA, USA.,Faculty of Physical Education, University of Brasília, Distrito Federal, Brazil
| | - Jahyun Kim
- Department of Kinesiology, Iowa State University, Ames, IA, USA
| | - Lauro C Vianna
- Faculty of Physical Education, University of Brasília, Distrito Federal, Brazil
| | - James A Lang
- Department of Kinesiology, Iowa State University, Ames, IA, USA
| |
Collapse
|
85
|
Hausenloy DJ, Bøtker HE. Why did remote ischaemic conditioning not improve clinical outcomes in acute myocardial infarction in the CONDI-2/ERIC-PPCI trial? Cardiovasc Res 2020; 115:e161-e163. [PMID: 31621833 DOI: 10.1093/cvr/cvz242] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, UK.,The National Institute of Health Research University College, London Hospitals Biomedical Research Centre, Research & Development, Maple House 1st floor, 149 Tottenham Court Road, London, UK.,Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore.,National Heart Research Institute Singapore, National Heart Centre, 5 Hospital Drive, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, Singapore.,Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, 64849 Nuevo Leon, Mexico
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark
| |
Collapse
|
86
|
Sawashita Y, Hirata N, Yoshikawa Y, Terada H, Tokinaga Y, Yamakage M. Remote ischemic preconditioning reduces myocardial ischemia-reperfusion injury through unacylated ghrelin-induced activation of the JAK/STAT pathway. Basic Res Cardiol 2020; 115:50. [PMID: 32607622 DOI: 10.1007/s00395-020-0809-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023]
Abstract
Remote ischemic preconditioning (RIPC) offers cardioprotection against myocardial ischemia-reperfusion injury. The humoral factors involved in RIPC that are released from parasympathetically innervated organs have not been identified. Previous studies showed that ghrelin, a hormone released from the stomach, is associated with cardioprotection. However, it is unknown whether or not ghrelin is involved in the mechanism of RIPC. This study aimed to determine whether ghrelin serves as one of the humoral factors in RIPC. RIPC group rats were subjected to three cycles of ischemia and reperfusion for 5 min in two limbs before left anterior descending (LAD) coronary artery ligation. Unacylated ghrelin (UAG) group rats were given 0.5 mcg/kg UAG intravenously 30 min before LAD ligation. Plasma levels of UAG in all groups were measured before and after RIPC procedures and UAG administration. Additionally, JAK2/STAT3 pathway inhibitor (AG490) was injected in RIPC and UAG groups to investigate abolishment of the cardioprotection of RIPC and UAG. Plasma levels of UAG, infarct size and phosphorylation of STAT3 were compared in all groups. Infarct size was significantly reduced in RIPC and UAG groups, compared to the other groups. Plasma levels of UAG in RIPC and UAG groups were significantly increased after RIPC and UAG administration, respectively. The cardioprotective effects of RIPC and UAG were accompanied by an increase in phosphorylation of STAT3 and abolished by AG490. This study indicated that RIPC reduces myocardial ischemia and reperfusion injury through UAG-induced activation of JAK/STAT pathway. UAG may be one of the humoral factors involved in the cardioprotective effects of RIPC.
Collapse
Affiliation(s)
- Yasuaki Sawashita
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan.
| | - Naoyuki Hirata
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yusuke Yoshikawa
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Hirofumi Terada
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yasuyuki Tokinaga
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Michiaki Yamakage
- Department of Anesthesiology, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| |
Collapse
|
87
|
Shahi HA, Kadoguchi T, Shimada K, Fukao K, Matsushita S, Aikawa T, Ouchi S, Shiozawa T, Takahashi S, Sato-Okabayashi Y, Akita K, Isoda K, Miyazaki T, Daida H. Voluntary exercise and cardiac remodeling in a myocardial infarction model. Open Med (Wars) 2020; 15:545-555. [PMID: 33313409 PMCID: PMC7706131 DOI: 10.1515/med-2020-0109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/01/2020] [Accepted: 03/10/2020] [Indexed: 02/04/2023] Open
Abstract
We investigated the effects of voluntary exercise after myocardial infarction (MI) on cardiac function, remodeling, and inflammation. Male C57BL/6J mice were divided into the following four groups: sedentary + sham (Sed-Sh), sedentary + MI (Sed-MI), exercise + sham (Ex-Sh), and exercise + MI (Ex-MI). MI induction was performed by ligation of the left coronary artery. Exercise consisting of voluntary wheel running started after the operation and continued for 4 weeks. The Ex-MI mice had significantly increased cardiac function compared with the Sed-MI mice. The Ex-MI mice showed significantly reduced expression levels of tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-10 in the infarcted area of the left ventricle compared with the Sed-MI mice. In the Ex-MI mice, the expression levels of fibrosis-related genes including collagen I and III were decreased compared to the Sed-MI mice, and the expression levels of IL-1β, IL-6, follistatin-like 1, fibroblast growth factor 21, and mitochondrial function-related genes were significantly elevated in skeletal muscle compared with the Sed mice. The plasma levels of IL-6 were also significantly elevated in the Ex-MI group compared with the Sed-MI groups. These findings suggest that voluntary exercise after MI may improve in cardiac remodeling associated with anti-inflammatory effects in the myocardium and myokine production in the skeletal muscles.
Collapse
Affiliation(s)
- Hamad Al Shahi
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoyasu Kadoguchi
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazunori Shimada
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kosuke Fukao
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Satoshi Matsushita
- Department of Cardiovascular Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tatsuro Aikawa
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shohei Ouchi
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoyuki Shiozawa
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shuhei Takahashi
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yayoi Sato-Okabayashi
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Akita
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kikuo Isoda
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tetsuro Miyazaki
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Faculty of Health Science, Juntendo University, Tokyo, Japan
| |
Collapse
|
88
|
Kane WJ, Hassinger TE, Elwood NR, Dietch ZC, Krebs ED, Popovsky KA, Hedrick TL, Sawyer RG. Fever Is Associated with Reduced Mortality in Trauma and Surgical Intensive Care Unit-Acquired Infections. Surg Infect (Larchmt) 2020; 22:174-181. [PMID: 32379549 DOI: 10.1089/sur.2019.352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Fever is a common response to both infectious and non-infectious physiologic insults in the critically ill, and in certain populations it appears to be protective. Fever is particularly common in trauma patients, and even more so in those with infections. The relationship between fever, trauma status, and mortality in patients with an infection is unclear. Patients and Methods: A review of a prospectively maintained institutional database over a 17-year period was performed. Surgical and trauma intensive care unit (ICU) patients with a nosocomial infection were extracted to compare in-hospital mortality among trauma and non-trauma patients with and without fever. Univariable analyses compared patient and infection characteristics between trauma and non-trauma patients. A multivariable logistic regression model was created to identify predictors of in-hospital mortality, with a focus on fever and trauma status. Results: Nine hundred forty-one trauma patients and 1,449 non-trauma patients with ICU-acquired infections were identified. Trauma patients were younger (48 vs. 59, p < 0.001), more likely to be male (73% vs. 56%, p < 0.001), more likely to require blood transfusion (74% vs. 47%, p < 0.001), had lower Acute Physiology and Chronic Health Evaluation (APACHE) II scores (18 vs. 19, p = 0.02), and had lower rates of comorbidities. Trauma patients were more likely to develop a fever (72% vs. 43%, p < 0.001) and had lower in-hospital mortality (9.6% vs. 22.6%, p < 0.001). In multivariable analysis, non-trauma patients with fever had a lower odds of mortality compared with non-trauma patients without fever (odds ratio [OR] 0.63, p = 0.004). Trauma patients with fever had the lowest odds ratio for mortality when compared to non-trauma patients without fever (OR 0.25, p < 0.001). Conclusions: In this large cohort of trauma and surgical ICU patients with ICU-acquired infections, fever was associated with a lower odds of mortality in both trauma and non-trauma patients. Further investigation is needed to determine the mechanisms behind the interplay between trauma status, fever, and mortality.
Collapse
Affiliation(s)
- William J Kane
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Taryn E Hassinger
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Nathan R Elwood
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Zachary C Dietch
- Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Elizabeth D Krebs
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | | | - Traci L Hedrick
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Robert G Sawyer
- Department of Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan, USA
| |
Collapse
|
89
|
Honkanen HP, Mustonen C, Herajärvi J, Tuominen H, Starck T, Kallio M, Kiviluoma K, Anttila V, Juvonen T. Remote Ischemic Preconditioning in Spinal Cord Protection: A Surviving Porcine Study. Semin Thorac Cardiovasc Surg 2020; 32:788-796. [PMID: 32380237 DOI: 10.1053/j.semtcvs.2020.03.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 11/11/2022]
Abstract
Surgical repair of thoracic aorta can compromise blood flow of the spinal cord. To mitigate spinal cord ischemia (SCI) additional protection methods are needed. In experimental studies remote ischemic preconditioning (RIPC) has proven to be an effective method of protecting organs from ischemia. The aim of the study was to assess efficacy of RIPC in spinal cord protection in a chronic porcine model. Sixteen piglets were assigned into the RIPC group (8) and the control group (8). RIPC was performed using blood pressure cuff in a 5-minute ischemia followed by a 5-minute reperfusion repeating cycles 4 times. The left subclavian artery and all segmental arteries above diaphragm were ligated at 5-minute intervals to accomplish SCI. The follow-up comprised a 4-hour intensive monitoring and a 7-day recovery phase. Blood samples were obtained, motor-evoked potentials and near-infrared spectroscopy (NIRS) of longitudinal back muscles were measured. Paraplegia was assessed every day postoperatively. Histopathological analysis of the spinal cord was performed after 7 days. NIRS values 4 hours after SCI were higher in the RIPC group, 45.5 (44.5-47.0), than in the control group, 41.5 (40.5-44.0) (P = 0.042). Nadir value of NIRS was 43.4 (39.3-46.0) in the RIPC group and 38.9 (38.-40.0) in the control group (P = 0.014). On the first postoperative day the RIPC group reached modified Tarlov score of 3 (2-3) vs 2 (1-2) in the control group (P = 0.024). RIPC hastens the recovery from SCI during the first postoperative day.
Collapse
Affiliation(s)
- Hannu-Pekka Honkanen
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Medical Research Center, Oulu, Finland.
| | - Caius Mustonen
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Medical Research Center, Oulu, Finland
| | - Johanna Herajärvi
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Medical Research Center, Oulu, Finland
| | - Hannu Tuominen
- Department of Pathology, Oulu University Hospital, Oulu, Finland
| | - Tuomo Starck
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland; Department of Neurophysiology, Oulu University Hospital, Oulu, Finland
| | - Mika Kallio
- Department of Neurophysiology, Oulu University Hospital, Oulu, Finland
| | - Kai Kiviluoma
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Medical Research Center, Oulu, Finland
| | - Vesa Anttila
- Heart Center, University of Turku and Turku University Hospital, Turku, Finland
| | - Tatu Juvonen
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Medical Research Center, Oulu, Finland; Department of Cardiac Surgery, Heart and Lung Center, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
90
|
Zhang XY, Huang Z, Li QJ, Zhong GQ, Meng JJ, Wang DX, Tu RH. Ischemic postconditioning attenuates the inflammatory response in ischemia/reperfusion myocardium by upregulating miR‑499 and inhibiting TLR2 activation. Mol Med Rep 2020; 22:209-218. [PMID: 32377693 PMCID: PMC7248531 DOI: 10.3892/mmr.2020.11104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/25/2020] [Indexed: 12/18/2022] Open
Abstract
Toll-like receptor 2 (TLR2)-mediated myocardial inflammation serves an important role in promoting myocardial ischemic/reperfusion (I/R) injury. Previous studies have shown that miR-499 is critical for cardioprotection after ischemic postconditioning (IPostC). Therefore, the present study evaluated the protective effect of IPostC on the myocardium by inhibiting TLR2, and also assessed the involvement of microRNA (miR)-499. Rat hearts were subjected to 30 min of ischemia and 2 h of reperfusion. The IPostC was 3 cycles of 30 sec of reperfusion and 30 sec of re-occlusion prior to reperfusion. In total, 90 rats were randomly divided into six groups (n=15 per group): Sham; I/R; IPostC; miR-499 negative control adeno-associated virus (AAV) vectors + IPostC; miR-499 inhibitor AAV vectors + IPostC; and miR-499 mimic AAV vectors + IPostC. It was identified that IPostC significantly decreased the I/R-induced cardiomyocyte apoptotic index (29.4±2.03% in IPostC vs. 42.64±2.27% in I/R; P<0.05) and myocardial infarct size (48.53±2.49% in IPostC vs. 66.52±3.1% in I/R; P<0.05). Moreover, these beneficial effects were accompanied by increased miR-499 expression levels (as demonstrated by reverse transcription-quantitative PCR) in the myocardial tissue and decreased TLR2, protein kinase C (PKC), interleukin (IL)-1β and IL-6 expression levels (as demonstrated by western blotting and ELISA) in the myocardium and serum. The results indicated that IPostC + miR-499 mimics significantly inhibited inflammation and the PKC signaling pathway and enhanced the anti-inflammatory and anti-apoptotic effects of IPostC. However, IPostC + miR-499 inhibitors had the opposite effect. Therefore, it was speculated that IPostC may have a miR-499-dependent cardioprotective effect. The present results suggested that miR-499 may be involved in IPostC-mediated ischemic cardioprotection, which may occur via local and systemic TLR2 inhibition, subsequent inhibition of the PKC signaling pathway and a decrease in inflammatory cytokine release, including IL-1β and IL-6. Moreover, these effects will ultimately lead to a decrease in the myocardial apoptotic index and myocardial infarct size via the induction of the anti-apoptotic protein Bcl-2, and inhibition of the pro-apoptotic protein Bax in myocardium.
Collapse
Affiliation(s)
- Xin-Yue Zhang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zheng Huang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qing-Jie Li
- Department of Cardiology, Second Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guo-Qiang Zhong
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jian-Jun Meng
- Department of Geriatric Health Care Center, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Dong-Xiao Wang
- Department of Cardiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rong-Hui Tu
- Guangxi Key Laboratory of Precision Medicine in Cardio‑Cerebrovascular Diseases Control and Prevention, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
91
|
Just J, Yan Y, Farup J, Sieljacks P, Sloth M, Venø M, Gu T, de Paoli FV, Nyengaard JR, Bæk R, Jørgensen MM, Kjems J, Vissing K, Drasbek KR. Blood flow-restricted resistance exercise alters the surface profile, miRNA cargo and functional impact of circulating extracellular vesicles. Sci Rep 2020; 10:5835. [PMID: 32245988 PMCID: PMC7125173 DOI: 10.1038/s41598-020-62456-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/10/2020] [Indexed: 01/12/2023] Open
Abstract
Ischemic exercise conducted as low-load blood flow restricted resistance exercise (BFRE) can lead to muscle remodelling and promote muscle growth, possibly through activation of muscle precursor cells. Cell activation can be triggered by blood borne extracellular vesicles (EVs) as these nano-sized particles are involved in long distance signalling. In this study, EVs isolated from plasma of healthy human subjects performing a single bout of BFRE were investigated for their change in EV surface profiles and miRNA cargos as well as their impact on skeletal muscle precursor cell proliferation. We found that after BFRE, five EV surface markers and 12 miRNAs were significantly altered. Furthermore, target prediction and functional enrichment analysis of the miRNAs revealed several target genes that are associated to biological pathways involved in skeletal muscle protein turnover. Interestingly, EVs from BFRE plasma increased the proliferation of muscle precursor cells. In addition, alterations in surface markers and miRNAs indicated that the combination of exercise and ischemic conditioning during BFRE can stimulate blood cells to release EVs. These results support that BFRE promotes EV release to engage in muscle remodelling and/or growth processes.
Collapse
Affiliation(s)
- Jesper Just
- Center of Functionally Integrative Neuroscience, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Yan Yan
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Jean Farup
- Research laboratory for Biochemical Pathology, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Dept of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Peter Sieljacks
- Section for Sport Science, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Mette Sloth
- Center of Functionally Integrative Neuroscience, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Morten Venø
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Tingting Gu
- Center of Functionally Integrative Neuroscience, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Jens Randel Nyengaard
- Dept of Clinical Medicine, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Rikke Bæk
- Dept of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Malene Møller Jørgensen
- Dept of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark.,Dept of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark.,Dept of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Kristian Vissing
- Section for Sport Science, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Kim Ryun Drasbek
- Center of Functionally Integrative Neuroscience, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
92
|
Nazari-Shafti TZ, Exarchos V, Biefer HRC, Cesarovic N, Meyborg H, Falk V, Emmert MY. MicroRNA Mediated Cardioprotection - Is There a Path to Clinical Translation? Front Bioeng Biotechnol 2020; 8:149. [PMID: 32266222 PMCID: PMC7099408 DOI: 10.3389/fbioe.2020.00149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
In the past 20 years, there have been several approaches to achieve cardioprotection or cardiac regeneration using a vast variety of cell therapies and remote ischemic pre-conditioning (RIPC). To date, substantial proof that either cell therapy or RIPC has the potential for clinically relevant cardiac repair or regeneration of cardiac tissue is still pending. Preclinical trials indicate that the secretome of cells in situ (during RIPC) as well as of transplanted cells may exhibit cardioprotective properties in the acute setting of cardiac injury. The secretome generally consists of cell-specific cytokines and extracellular vesicles (EVs) containing microRNAs (miRNAs). It is currently hypothesized that a subset of known miRNAs play a crucial part in the facilitation of cardioprotective effects. miRNAs are small non-coding RNA molecules that inhibit post-transcriptional translation of messenger RNAs (mRNAs) and play an important role in gene translation regulation. It is also known that one miRNAs usually targets multiple mRNAs. This makes predictability of pharmacokinetics and mechanism of action very difficult and could in part explain the inferior performance of various progenitor cells in clinical studies. Identification of miRNAs involved in cardioprotection and remodeling, the composition of miRNA profiles, and the exact mechanism of action are important to the design of future cell-based but also cell-free cardioprotective therapeutics. This review will give a description of miRNA with cardioprotective properties and a current overview on known mechanism of action and potential missing links. Additionally, we will give an outlook on the potential for clinical translation of miRNAs in the setting of myocardial infarction and heart failure.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Deutsches Zentrum für Herz-und Kreislauferkrankungen, Berlin, Germany
| | - Vasileios Exarchos
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Héctor Rodriguez Cetina Biefer
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Clinic for Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nikola Cesarovic
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Heike Meyborg
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany
| | - Volkmar Falk
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Deutsches Zentrum für Herz-und Kreislauferkrankungen, Berlin, Germany.,Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian Y Emmert
- Department for Cardiovascular and Thoracic Surgery, German Heart Center Berlin, Berlin, Germany.,Deutsches Zentrum für Herz-und Kreislauferkrankungen, Berlin, Germany.,Clinic for Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
93
|
He Q, Wang F, Ryan TD, Chalasani M, Redington AN. Repeated Remote Ischemic Conditioning Reduces Doxorubicin-Induced Cardiotoxicity. JACC: CARDIOONCOLOGY 2020; 2:41-52. [PMID: 34396208 PMCID: PMC8352345 DOI: 10.1016/j.jaccao.2020.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 02/06/2023]
Abstract
Objectives This study investigated the cardioprotective effect of repeated remote ischemic preconditioning (rRIC) on doxorubicin-induced cardiotoxicity in mice. Background Doxorubicin is an effective chemotherapeutic agent for a wide range of tumor types but its use and dosing are limited by acute and chronic cardiotoxicity. Remote ischemic conditioning (RIC) is cardioprotective in multiple cardiovascular injury models, but the effectiveness of rRIC in doxorubicin-induced cardiotoxicity has not been fully elucidated. Methods rRIC was performed on mice before and after doxorubicin administration. Cardiac function was assessed by echocardiography and myocardial biology was tested by molecular approaches. Results Doxorubicin administration induced acute cardiotoxicity, as indicated by reduced cardiac function, reduced myocyte cross-section area and increased extracellular collagen deposition, increased circulating cardiac muscle damage markers, and decreased heart weight. Doxorubicin also adversely affected other organs, including the kidney, liver, and spleen, as evaluated by circulating markers or organ weight loss. rRIC not only abrogated doxorubicin-induced cardiotoxicity (left ventricular ejection fraction, doxorubicin 47.5 ± 1.1%, doxorubicin + rRIC 51.6 ± 0.7%, p = 0.017), but also was associated with multiorgan protection. Within the myocardium, rRIC attenuated doxorubicin-induced cardiomyocyte apoptosis, reduced inflammation, and increased autophagy signaling. Conclusions rRIC may be a promising approach to reduce doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Quan He
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Fangfei Wang
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Thomas D Ryan
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Meghana Chalasani
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew N Redington
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
94
|
Hausenloy DJ, Ng CT, Chong JH. Repeated Remote Ischemic Conditioning Protects Against Doxorubicin Cardiotoxicity: Never Too Much of a Good Thing. JACC CardioOncol 2020; 2:53-55. [PMID: 34396209 PMCID: PMC8352202 DOI: 10.1016/j.jaccao.2020.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung City, Taiwan
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
| | - Choon Ta Ng
- Department of Cardiology, National Heart Centre Singapore, Singapore
| | - Jun Hua Chong
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- Department of Cardiology, National Heart Centre Singapore, Singapore
| |
Collapse
|
95
|
MiRNA-Mediated Mechanisms of Cardiac Protection in Ischemic and Remote Ischemic Preconditioning-A Qualitative Systematic Review. Shock 2020; 51:44-51. [PMID: 29642230 DOI: 10.1097/shk.0000000000001156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Ischemic preconditioning (IPC) and remote ischemic preconditioning (RIPC) protect myocardial tissue against subsequent ischemia and reperfusion injury (IRI) and have a high potential to improve patient outcome. The mediators and mechanisms of protection through IPC and RIPC remain largely unknown, but micro-RNAs (miRNAs) are promising candidates. METHODS Systematic review of Medline and Embase databases for biomedical scientific literature. RESULTS A total of 26 relevant publications (21 full-text original articles and 5 conference abstracts) were identified, 8 describing cell culture experiments, 14 animal experiments, and 4 randomized clinical trials in humans. Most commonly reported miRNAs with differential expression between preconditioned and control groups include miR-1, miR-21, and miR-144. Experimental designs and procedures differ widely, thereby limiting the potential to compare results between studies. Two of the four RCTs did not find any differentially expressed miRNAs. CONCLUSIONS Results from RCTs should feed back into basic research and focused studies confirming or rejecting hypotheses generated by these RCTs are needed.
Collapse
|
96
|
Late Phases of Cardioprotection During Remote Ischemic Preconditioning and Adenosine Preconditioning Involve Activation of Neurogenic Pathway. J Cardiovasc Pharmacol 2020; 73:63-69. [PMID: 30422893 DOI: 10.1097/fjc.0000000000000634] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The role of the neurogenic pathway in early phases of cardioprotection during remote ischemic preconditioning (RIPC) and adenosine preconditioning is reported. AIM This study was designed to explore the involvement of the neurogenic pathway in late phases of cardioprotection during RIPC and adenosine preconditioning. MATERIAL AND METHODS Fifty-four Wistar rats were used and divided into 9 experimental groups. RIPC was induced by tying the blood pressure cuff around the hind limb and subjecting to 4 cycles of inflation and deflation of 5 minutes each. In early RIPC, the heart was isolated immediately after the last episode of RIPC, whereas in late RIPC, the heart was isolated 24 hours after the last cycle of RIPC. In a similar way, adenosine preconditioning was instituted in early and late phases by either isolating the heart 40 minutes or 24 hours after adenosine (4 mg/kg, intraperitoneally [i.p.]) administration. Isolated hearts were subjected to ischemia-reperfusion (I/R) injury on the Langendorff's system. RESULTS Both early and late phases of RIPC and adenosine preconditioning significantly abrogated I/R-induced myocardial injury in terms of decrease in the release of lactate dehydrogenase, creatine kinase, and decrease in infarct size. Pretreatment with hexamethonium, a ganglion blocker (20 mg/kg, i.p.), significantly abolished the cardioprotective effects of both early and late phases of RIPC and adenosine preconditioning. CONCLUSION Apart from the involvement of the neurogenic pathway in the early phases, there is a critical role of the neurogenic pathway in the late phase of cardioprotection during RIPC and adenosine preconditioning.
Collapse
|
97
|
Davila S, Jaquiss RD. Commentary: Remote Ischemic Preconditioning-Too Good to Be True? Semin Thorac Cardiovasc Surg 2020; 32:325-326. [PMID: 32057967 DOI: 10.1053/j.semtcvs.2020.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/04/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Samuel Davila
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center & Children's Health System of Texas, Dallas, Texas
| | - Robert Db Jaquiss
- Division of Pediatric Cardiothoracic Surgery, Department of Thoracic and Cardiovascular Surgery, University of Texas Southwestern Medical Center & Children's Health System of Texas, Dallas, Texas.
| |
Collapse
|
98
|
Chen K, Yu J, Wang Q, Wu L, Liu X, Wong GTC, Lu Y. The timing of propofol administration affects the effectiveness of remote ischemic preconditioning induced cardioprotection in rats. J Cell Biochem 2020; 121:4535-4541. [PMID: 32030809 DOI: 10.1002/jcb.29671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
The cardioprotection of remote ischemic preconditioning (RIPC) is abolished under propofol maintained anesthesia. Transient receptor potential vanilloid 1 (TRPV1) channel is present in the heart, and its activation could induce cardioprotection. Therefore, we tested whether the anesthetic propofol administration phase interfered with the RIPC-induced cardioprotection, and RIPC-induced cardioprotection via the cardiac TRPV1 channel. Male Sprague-Dawley rats were subjected to myocardial 30 minutes of ischemia followed by 2 hours of reperfusion. RIPC consisted of three cycles of 5-minute ischemia/reperfusion applied to a hindlimb. Propofol infusion at 12 mg/kg/h was commenced either at 10 minutes before the start of RIPC in the P-pre + RIPC group, or immediately after myocardial ischemia at the onset of reperfusion (P-post + RIPC) while performing RIPC. These two propofol infusion regimes were applied to another two grou bs without RIPC (P-pre and P-post groups). Infarct size (IS) was assessed by triphenyltetrazolium staining. Heart TRPV1 expression was detected by Western blot and immunofluorescence. RIPC significantly reduced myocardial IS compared with the control group (36.7 ± 3% versus 57.2 ± 4%; P < .01). When propofol was started before RIPC, the IS sparing effect of RIPC was completely abolished. However, propofol infusion starting immediately after myocardial ischemia did not affect RIPC-induced cardioprotection. TRPV1 expression significant increase after RIPC, then propofol inhibited the TRPV1 activation of RIPC if given before RIPC but not after. Our results suggest that the timing of propofol administration is critical to preserve the cardioprotection of RIPC. Propofol might cancel RIPC-induced cardioprotection via the cardiac TRPV1 receptor.
Collapse
Affiliation(s)
- Ke Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junma Yu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China.,Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiuyue Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lining Wu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | | | - Yao Lu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
99
|
Remote Ischemic Preconditioning in Microsurgical Head and Neck Reconstruction: A Randomized Controlled Trial. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e2591. [PMID: 32095401 PMCID: PMC7015612 DOI: 10.1097/gox.0000000000002591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 01/19/2023]
Abstract
Background: The free flap failure rate is 5% in head and neck microsurgical reconstruction, and ischemia–reperfusion injury is an important mechanism behind this failure rate. Remote ischemic preconditioning (RIPC) is a recent intervention targeting ischemia–reperfusion injury. The aim of the present study was to investigate if RIPC improved clinical outcomes in microsurgical reconstruction. Methods: Head and neck cancer patients undergoing tumor resection and microsurgical reconstruction were included in a randomized controlled trial. Patients were randomized (1:1) to RIPC or sham intervention administered intraoperatively just before transfer of the free flap. RIPC was administered by four 5-minute periods of upper extremity occlusion and reperfusion. Clinical data were prospectively collected in the perioperative period and at follow-up on postoperative days 30 and 90. Intention-to-treat analysis was performed. Results: Sixty patients were randomized to RIPC (n = 30) or sham intervention (n = 30). All patients received allocated intervention. No patients were lost to follow up. At 30-day follow-up, flap failure occurred in 7% of RIPC patients (n = 2) and 3% of sham patients (n = 1) with the relative risk and 95% confidence interval 2.0 [0.2;20.9], P = 1.0. The rate of pedicle thrombosis was 10% (n = 3) in both groups with relative risk 1.0 [0.2;4.6], P = 1.0. The flap failure rate did not change at 90-day follow-up. Conclusions: RIPC is safe and feasible but does not affect clinical outcomes in head and neck cancer patients undergoing microsurgical reconstruction.
Collapse
|
100
|
Zhou H, Yang L, Wang G, Zhang C, Fang Z, Lei G, Shi S, Li J. Remote Ischemic Preconditioning Prevents Postoperative Acute Kidney Injury After Open Total Aortic Arch Replacement: A Double-Blind, Randomized, Sham-Controlled Trial. Anesth Analg 2020; 129:287-293. [PMID: 30896603 DOI: 10.1213/ane.0000000000004127] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Acute kidney injury is a common complication after open total aortic arch replacement but lacks effective preventive strategies. Remote ischemic preconditioning has controversial results of its benefit to the kidney and may perform better in high-risk patients of acute kidney injury. We investigated whether remote ischemic preconditioning would prevent postoperative acute kidney injury after open total aortic arch replacement. METHODS We enrolled 130 patients scheduled for open total aortic arch replacement and randomized them to receive either remote ischemic preconditioning (4 cycles of 5-minute right upper limb ischemia and 5-minute reperfusion) or sham preconditioning (4 cycles of 5-minute right upper limb pseudo ischemia and 5-minute reperfusion), both via blood pressure cuff inflation and deflation. The primary end point was the incidence of acute kidney injury within 7 days after the surgery defined by the Kidney Disease: Improving Global Outcomes criteria. Secondary end point included short-term clinical outcomes. RESULTS Significantly fewer patients developed postoperative acute kidney injury with remote ischemic preconditioning compared with sham (55.4% vs 73.8%; absolute risk reduction, 18.5%; 95% CI, 2.3%-34.6%; P = .028). Remote ischemic preconditioning significantly reduced acute kidney injury stage II-III (10.8% vs 35.4%; P = .001). Remote ischemic preconditioning shortened the mechanical ventilation duration (18 hours [interquartile range, 14-33] versus 25 hours [interquartile range, 17-48]; P = .01), whereas no significant differences were observed between groups in other secondary outcomes. CONCLUSIONS Remote ischemic preconditioning prevented acute kidney injury after open total aortic arch replacement, especially severe acute kidney injury and shortened mechanical ventilation duration. The observed renoprotective effects of remote ischemic preconditioning require further investigation in both clinical research and the underlying mechanism.
Collapse
Affiliation(s)
- Hui Zhou
- From the Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lijing Yang
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guyan Wang
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Congya Zhang
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongrong Fang
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guiyu Lei
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng Shi
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Li
- Department of Anesthesiology, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|