1
|
Li Y, Chen Y, Yu P, Zhang D, Tang X, Zhu Z, Xiao F, Deng W, Liu Y, Tan Z, Zhang J, Yu S. Mild therapeutic hypothermic protection activates the PI3K/AKT signaling pathway to inhibit TRPM7 and suppress ferroptosis induced by myocardial ischemia‑reperfusion injury. Mol Med Rep 2024; 30:220. [PMID: 39364741 PMCID: PMC11462392 DOI: 10.3892/mmr.2024.13345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024] Open
Abstract
The present study aimed to investigate the role of PI3K‑mediated ferroptosis signaling induced by mild therapeutic hypothermia (MTH), which was defined as a temperature of 34˚C, in protecting against myocardial ischemia-reperfusion (I/R) injury (MIRI). To meet this aim, H9C2 cells underwent hypoxia‑reperfusion (H/R) and/or MTH. The MTT assay was used to assess cell viability, cytotoxicity was measured using a lactate dehydrogenase cytotoxicity assay, and Annexin V‑FITC/PI flow cytometric analysis was used to analyze early and late cell apoptosis. In addition, 84 healthy adult male Sprague‑Dawley rats were randomly divided into seven groups (n=12), and underwent I/R and various treatments. Hemodynamics were monitored, and the levels of myocardial injury marker enzymes and oxidative stress markers in myocardial tissue were measured using ELISA. The expression levels of PI3K, AKT, transient receptor potential cation channel subfamily M member 7 (TRPM7), glutathione peroxidase 4 (GPX4) and acyl‑CoA synthetase long chain family member 4 (ACSL4) in animals and cells were measured using western blot analysis. These experiments revealed that MTH could effectively reduce myocardial infarct size, improve hemodynamic performance following MIRI and suppress myocardial apoptosis, thereby contributing to the recovery from H/R injury. Mechanistically, MTH was revealed to be able to activate the PI3K/AKT signaling pathway in cells, upregulating GPX4, and downregulating the expression levels of TRPM7 and ACSL4. Treatment with 2‑aminoethoxydiphenyl borate (an inhibitor of TRPM7) could further strengthen the myocardial protective effects of MTH, whereas treatment with erastin (promoter of ferroptosis) and wortmannin (inhibitor of PI3K) led to the effective elimination of the myocardial protective effects of MTH. Compared with in the I/R group, the PI3K/AKT activation level and the expression levels of GPX4 were both significantly increased, whereas the expression levels of TRPM7 and ACSL4 were significantly decreased in the I/R + MTH group. Taken together, the results of the present study indicated that MTH may activate the PI3K/AKT signaling pathway to inhibit TRPM7 and suppress ferroptosis induced by MIRI.
Collapse
Affiliation(s)
- Yaqi Li
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Department of Anesthesiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, P.R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Xiaoyi Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wei Deng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhaoying Tan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shuchun Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2
|
Berg J, Jablonowski R, Nordlund D, Ryd D, Heiberg E, Carlsson M, Arheden H. Mild hypothermia attenuates ischaemia/reperfusion injury: insights from serial non-invasive pressure-volume loops. Cardiovasc Res 2023; 119:2230-2243. [PMID: 36734080 PMCID: PMC10578916 DOI: 10.1093/cvr/cvad028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 02/04/2023] Open
Abstract
AIMS Mild hypothermia, 32-35°C, reduces infarct size in experimental studies, potentially mediating reperfusion injuries, but human trials have been ambiguous. To elucidate the cardioprotective mechanisms of mild hypothermia, we analysed cardiac performance in a porcine model of ischaemia/reperfusion, with serial cardiovascular magnetic resonance (CMR) imaging throughout 1 week using non-invasive pressure-volume (PV) loops. METHODS AND RESULTS Normothermia and Hypothermia group sessions (n = 7 + 7 pigs, non-random allocation) were imaged with Cardiovascular magnetic resonance (CMR) at baseline and subjected to 40 min of normothermic ischaemia by catheter intervention. Thereafter, the Hypothermia group was rapidly cooled (mean 34.5°C) for 5 min before reperfusion. Additional CMR sessions at 2 h, 24 h, and 7 days acquired ventricular volumes and ischaemic injuries (unblinded analysis). Stroke volume (SV: -24%; P = 0.029; Friedmans test) and ejection fraction (EF: -20%; P = 0.068) were notably reduced at 24 h in the Normothermia group compared with baseline. In contrast, the decreases were ameliorated in the Hypothermia group (SV: -6%; P = 0.77; EF: -6%; P = 0.13). Mean arterial pressure remained stable in Normothermic animals (-3%, P = 0.77) but dropped 2 h post-reperfusion in hypothermic animals (-18%, P = 0.007). Both groups experienced a decrease and partial recovery pattern for PV loop-derived variables over 1 week, but the adverse effects tended to attenuate in the Hypothermia group. Infarct sizes were 10 ± 8% in Hypothermic and 15 ± 8% in Normothermic animals (P = 0.32). Analysis of covariance at 24 h indicated that hypothermia has cardioprotective properties incremental to reducing infarct size, such as higher external power (P = 0.061) and lower arterial elastance (P = 0.015). CONCLUSION Using non-invasive PV loops by CMR, we observed that mild hypothermia at reperfusion alleviates the heart's work after ischaemia/reperfusion injuries during the first week and preserves short-term cardiac performance. This hypothesis-generating study suggests hypothermia to have cardioprotective properties, incremental to reducing infarct size. The primary cardioprotective mechanism was likely an afterload reduction acutely unloading the left ventricle.
Collapse
Affiliation(s)
- Jonathan Berg
- Clinical Physiology, Department of Clinical Sciences LundFaculty of Medicine, Lund University, Box 117 221 00 Lund, Sweden
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
- Syntach AB, Lund, Sweden
| | - Robert Jablonowski
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
| | - David Nordlund
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
| | - Daniel Ryd
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
| | - Einar Heiberg
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
| | - Marcus Carlsson
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
| | - Håkan Arheden
- Skåne University Hospital, Carl-Bertil Laurells gata 9, 214 28 Malmö, Sweden
| |
Collapse
|
3
|
Belur AD, Sedhai YR, Truesdell AG, Khanna AK, Mishkin JD, Belford PM, Zhao DX, Vallabhajosyula S. Targeted Temperature Management in Cardiac Arrest: An Updated Narrative Review. Cardiol Ther 2023; 12:65-84. [PMID: 36527676 PMCID: PMC9986171 DOI: 10.1007/s40119-022-00292-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
The established benefits of cooling along with development of sophisticated methods to safely and precisely induce, maintain, monitor, and reverse hypothermia have led to the development of targeted temperature management (TTM). Early trials in human subjects showed that hypothermia conferred better neurological outcomes when compared to normothermia among survivors of cardiac arrest, leading to guidelines recommending targeted hypothermia in this patient population. Multiple studies have sought to explore and compare the benefit of hypothermia in various subgroups of patients, such as survivors of out-of-hospital cardiac arrest versus in-hospital cardiac arrest, and survivors of an initial shockable versus non-shockable rhythm. Larger and more recent trials have shown no statistically significant difference in neurological outcomes between patients with targeted hypothermia and targeted normothermia; further, aggressive cooling is associated with a higher incidence of multiple systemic complications. Based on this data, temporal trends have leaned towards using a lenient temperature target in more recent times. Current guidelines recommend selecting and maintaining a constant target temperature between 32 and 36 °C for those patients in whom TTM is used (strong recommendation, moderate-quality evidence), as soon as possible after return of spontaneous circulation is achieved and airway, breathing (including mechanical ventilation), and circulation are stabilized. The comparative benefit of lower (32-34 °C) versus higher (36 °C) temperatures remains unknown, and further research may help elucidate this. Any survivor of cardiac arrest who is comatose (defined as unarousable unresponsiveness to external stimuli) should be considered as a candidate for TTM regardless of the initial presenting rhythm, and the decision to opt for targeted hypothermia versus targeted normothermia should be made on a case-by-case basis.
Collapse
Affiliation(s)
- Agastya D Belur
- Division of Cardiology, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Yub Raj Sedhai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Kentucky College of Medicine, Bowling Green, KY, USA
| | | | - Ashish K Khanna
- Section of Critical Care Medicine, Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Outcomes Research Consortium, Cleveland, OH, USA.,Perioperative Outcomes and Informatics Collaborative (POIC), Winston-Salem, NC, USA
| | - Joseph D Mishkin
- Section of Advanced Heart Failure and Transplant Cardiology, Atrium Health Sanger Heart and Vascular Institute, Charlotte, NC, USA
| | - P Matthew Belford
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest School of Medicine, 306 Westwood Avenue, Suite 401, High Point, Winston-Salem, NC, 27262, USA
| | - David X Zhao
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest School of Medicine, 306 Westwood Avenue, Suite 401, High Point, Winston-Salem, NC, 27262, USA
| | - Saraschandra Vallabhajosyula
- Perioperative Outcomes and Informatics Collaborative (POIC), Winston-Salem, NC, USA. .,Section of Cardiovascular Medicine, Department of Medicine, Wake Forest School of Medicine, 306 Westwood Avenue, Suite 401, High Point, Winston-Salem, NC, 27262, USA. .,Department of Implementation Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
4
|
Xue Q, Zhang Q, Guo Z, Wu L, Chen Y, Chen Z, Yang K, Cao J. Therapeutic Hypothermia Inhibits Hypoxia-Induced Cardiomyocyte Apoptosis Via the MiR-483-3p/Cdk9 Axis. J Am Heart Assoc 2023; 12:e026160. [PMID: 36789845 PMCID: PMC10111479 DOI: 10.1161/jaha.122.026160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background Therapeutic hypothermia has a beneficial effect on cardiac function after acute myocardial infarction, but the exact mechanism is still unclear. Recent research has suggested that microRNAs participate in acute myocardial infarction to regulate cardiomyocyte survival. This study aimed to explore the ability of hypothermia-regulated microRNA-483-3p (miR-483-3p) to inhibit hypoxia-induced myocardial infarction. Methods and Results Primary cardiomyocytes were cultured under hypoxia at 32 °C to mimic therapeutic hypothermia, and the differentially expressed microRNAs were determined by RNA sequencing. Therapeutic hypothermia recovered hypoxia-induced increases in apoptosis, decreases in ATP levels, and decreases in miR-483-3p expression. Overexpression of miR-483-3p exhibited effects similar to those of therapeutic hypothermia on hypoxia in the treatment of cardiomyocytes to associate with maintaining the mitochondrial membrane potential, and cyclin-dependent kinase 9 (Cdk9) was identified as a target gene with downregulated expression by miR-483-3p. Knockdown of Cdk9 also promoted cardiac survival, ATP production, and mitochondrial membrane potential stability under hypoxia. In vivo, the expression of miR-483-3p and Cdk9 was tested in the cardiac tissue of the mice with acute myocardial infarction, and the expression of miR-483-3p decreased and Cdk9 increased in the region of myocardial infarction. However, miR-483-3p was overexpressed with lentivirus, which suppressed apoptosis, infarct size (miR-483-3p, 22.00±4.04% versus negative control, 28.57±5.44%, P<0.05), and Cdk9 expression to improve cardiac contractility. Conclusions MiR-483-3p antagonizes hypoxia, leading to cardiomyocyte injury by targeting Cdk9, which is a new mechanism of therapeutic hypothermia.
Collapse
Affiliation(s)
- Qiqi Xue
- Department of Geriatrics Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Qianru Zhang
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Zhenzhen Guo
- Department of Cardiovascular Medicine Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Liping Wu
- Department of Cardiac Imaging Center The First Affiliated Hospital, Auhui Medical University Hefei China
| | - Yafen Chen
- Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital, Fudan University Shanghai China
| | - Zhongli Chen
- State Key Laboratory of Cardiovascular Disease Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Ke Yang
- Department of Cardiovascular Medicine Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jiumei Cao
- Department of Geriatrics Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
5
|
Jung KT, Bapat A, Kim YK, Hucker WJ, Lee K. Therapeutic hypothermia for acute myocardial infarction: a narrative review of evidence from animal and clinical studies. Korean J Anesthesiol 2022; 75:216-230. [PMID: 35350095 PMCID: PMC9171548 DOI: 10.4097/kja.22156] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/21/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death from coronary heart disease and requires immediate reperfusion therapy with thrombolysis, primary percutaneous coronary intervention, or coronary artery bypass grafting. However, myocardial reperfusion therapy is often accompanied by cardiac ischemia/reperfusion (I/R) injury, which leads to myocardial injury with detrimental consequences. The causes of I/R injury are unclear, but are multifactorial, including free radicals, reactive oxygen species, calcium overload, mitochondria dysfunction, inflammation, and neutrophil-mediated vascular injury. Mild hypothermia has been introduced as one of the potential inhibitors of myocardial I/R injury. Although animal studies have demonstrated that mild hypothermia significantly reduces or delays I/R myocardium damage, human trials have not shown clinical benefits in acute MI (AMI). In addition, the practice of hypothermia treatment is increasing in various fields such as surgical anesthesia and intensive care units. Adequate sedation for anesthetic procedures and protection from body shivering has become essential during therapeutic hypothermia. Therefore, anesthesiologists should be aware of the effects of therapeutic hypothermia on the metabolism of anesthetic drugs. In this paper, we review the existing data on the use of therapeutic hypothermia for AMI in animal models and human clinical trials to better understand the discrepancy between perceived benefits in preclinical animal models and the absence thereof in clinical trials thus far.
Collapse
Affiliation(s)
- Ki Tae Jung
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology and Pain Medicine, College of Medicine and Medical School, Chosun University, Gwangju, Korea
| | - Aneesh Bapat
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cardiac Arrhythmia Service, Massachusetts General Hospital, Boston, MA, USA
| | - Young-Kug Kim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - William J. Hucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cardiac Arrhythmia Service, Massachusetts General Hospital, Boston, MA, USA
| | - Kichang Lee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cardiac Arrhythmia Service, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
6
|
Xu S, Miao H, Gong L, Feng L, Hou X, Zhou M, Shen H, Chen W. Effects of Different Hypothermia on the Results of Cardiopulmonary Resuscitation in a Cardiac Arrest Rat Model. DISEASE MARKERS 2022; 2022:2005616. [PMID: 35419118 PMCID: PMC9001110 DOI: 10.1155/2022/2005616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/02/2022] [Indexed: 11/17/2022]
Abstract
Objective To investigate the optimal temperature of hypothermia treatment in rats with cardiac arrest caused by ventricular fibrillation (VF) after the return of spontaneous circulation (ROSC). Methods A total of forty-eight male Sprague-Dawley rats were induced by VF through the guidewire with a maximum of 5 mA current and untreated for 8 min. Cardiopulmonary resuscitation (CPR) was performed for 8 min followed by defibrillation (DF). Resuscitated rats were then randomized into the normothermia (37°C) group, milder (35°C) group, mild (33°C) group, or moderate (28°C) group. Hypothermia was immediately induced with surface cooling. The target temperature was maintained for 4 h before rewarming to 37 ± 0.5°C. Moreover, at the end of the 4 h, a rat in each group was randomly selected to be sacrificed for the cerebral cortex electron microscopy observation (n = 1). The other resuscitated animals were observed for up to 72 h after ROSC (n = 7). Left ventricular ejection fraction (LVEF) and left ventricular end diastolic volume (LVEDV) were measured. Survival time, survival rate, and neurological deficit score (NDS) were recorded for 72 h. Results During hypothermia, higher LVEF was observed in the hypothermia groups when compared with normothermia group (35°C vs. 37°C, p < 0.05, 33°C and 28°C vs. 37°C, p < 0.01). Among the hypothermia groups, LVEF was higher in the 28°C group than that of 35°C (p < 0.05). However, both the heart rate (HR) (p < 0.01) and LVEDV (28°C vs. 35°C, p < 0.01, 28°C vs. 37°C and 33°C, p < 0.05) were lowest in the 28°C group when compared with the other groups. There were no significant differences of LVEF and LVEDV between the group 35°C and 33°C (p > 0.05). After rewarming, the LVEF of 35°C group was higher than that of group 37°C, 33°C, and 28°C (35°C vs. 37°C and 28°C, p < 0.01, 35°C vs. 33°C, p < 0.05). Group 35°C and 33°C resulted in longer survival (p < 0.01), higher survival rate (p < 0.01), and lower NDS (35°C vs. 37°C and 28°C, p < 0.01, 33°C vs. 37°C and 28°C, p < 0.05) compared with the group 37°C and 28°C. The extent of damage to cerebral cortex cells in group of 35°C and 33°C was lighter than that in group of 37°C and 28°C. The 35°C group spent less time in the process of cooling and rewarming than the group 33°C and 28°C (p < 0.01). Conclusions An almost equal protective effect of milder hypothermia (35°C) and mild hypothermia (33°C) in cardiac arrest (CA) rats was achieved with more predominant effect than moderate hypothermia (28°C) and normothermia (37°C). More importantly, shorter time spent in cooling and rewarming was required in the 35°C group, indicating its potential clinical application. These findings support the possible use of milder hypothermia (35°C) as a therapeutic agent for postresuscitation.
Collapse
Affiliation(s)
- Shaohua Xu
- Nankai University School of Medicine, Tianjin, China
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hui Miao
- The 3rd Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liming Gong
- Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Lijie Feng
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xuliang Hou
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Manhong Zhou
- Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Hong Shen
- Nankai University School of Medicine, Tianjin, China
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wei Chen
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
- The 3rd Medical Center of Chinese PLA General Hospital, Beijing, China
- Hainan Hospital of Chinese PLA General Hospital, Hainan, China
| |
Collapse
|
7
|
Nishi M, Ogata T, Kobayakawa K, Kobayakawa R, Matsuo T, Cannistraci CV, Tomita S, Taminishi S, Suga T, Kitani T, Higuchi Y, Sakamoto A, Tsuji Y, Soga T, Matoba S. Energy-sparing by 2-methyl-2-thiazoline protects heart from ischaemia/reperfusion injury. ESC Heart Fail 2021; 9:428-441. [PMID: 34854235 PMCID: PMC8787978 DOI: 10.1002/ehf2.13732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/13/2021] [Accepted: 11/11/2021] [Indexed: 11/06/2022] Open
Abstract
AIMS Cardiac ischaemia/reperfusion (I/R) injury remains a critical issue in the therapeutic management of ischaemic heart failure. Although mild hypothermia has a protective effect on cardiac I/R injury, more rapid and safe methods that can obtain similar results to hypothermia therapy are required. 2-Methyl-2-thiazoline (2MT), an innate fear inducer, causes mild hypothermia resulting in resistance to critical hypoxia in cutaneous or cerebral I/R injury. The aim of this study is to demonstrate the protective effect of systemically administered 2MT on cardiac I/R injury and to elucidate the mechanism underlying this effect. METHODS AND RESULTS A single subcutaneous injection of 2MT (50 mg/kg) was given prior to reperfusion of the I/R injured 10 week-old male mouse heart and its efficacy was evaluated 24 h after the ligation of the left anterior descending coronary artery. 2MT preserved left ventricular systolic function following I/R injury (ejection fraction, %: control 37.9 ± 6.7, 2MT 54.1 ± 6.4, P < 0.01). 2MT also decreased infarct size (infarct size/ischaemic area at risk, %: control 48.3 ± 12.1, 2MT 25.6 ± 4.2, P < 0.05) and serum cardiac troponin levels (ng/mL: control 8.9 ± 1.1, 2MT 1.9 ± 0.1, P < 0.01) after I/R. Moreover, 2MT reduced the oxidative stress-exposed area within the heart (%: control 25.3 ± 4.7, 2MT 10.8 ± 1.4, P < 0.01). These results were supported by microarray analysis of the mouse hearts. 2MT induced a transient, mild decrease in core body temperature (°C: -2.4 ± 1.4), which gradually recovered over several hours. Metabolome analysis of the mouse hearts suggested that 2MT minimized energy metabolism towards suppressing oxidative stress. Furthermore, 18F-fluorodeoxyglucose-positron emission tomography/computed tomography imaging revealed that 2MT reduced the activity of brown adipose tissue (standardized uptake value: control 24.3 ± 6.4, 2MT 18.4 ± 5.8, P < 0.05). 2MT also inhibited mitochondrial respiration and glycolysis in rat cardiomyoblasts. CONCLUSIONS We identified the cardioprotective effect of systemically administered 2MT on cardiac I/R injury by sparing energy metabolism with reversible hypothermia. Our results highlight the potential of drug-induced hypothermia therapy as an adjunct to coronary intervention in severe ischaemic heart disease.
Collapse
Affiliation(s)
- Masahiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Ko Kobayakawa
- Functional Neuroscience Lab, Kansai Medical University, Hirakata, Japan
| | - Reiko Kobayakawa
- Functional Neuroscience Lab, Kansai Medical University, Hirakata, Japan
| | - Tomohiko Matsuo
- Functional Neuroscience Lab, Kansai Medical University, Hirakata, Japan
| | - Carlo Vittorio Cannistraci
- Center for Complex Network Intelligence (CCNI), Tsinghua Laboratory of Brain and Intelligence (THBI), Department of Computer Science, Department of Biomedical Engineering, Tsinghua University, China.,Center for Systems Biology Dresden (CSBD), Dresden, Germany
| | - Shinya Tomita
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shunta Taminishi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takaomi Suga
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoya Kitani
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akira Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
8
|
Yamada KP, Kariya T, Aikawa T, Ishikawa K. Effects of Therapeutic Hypothermia on Normal and Ischemic Heart. Front Cardiovasc Med 2021; 8:642843. [PMID: 33659283 PMCID: PMC7919696 DOI: 10.3389/fcvm.2021.642843] [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: 12/16/2020] [Accepted: 01/21/2021] [Indexed: 12/24/2022] Open
Abstract
Therapeutic hypothermia has been used for treating brain injury after out-of-hospital cardiac arrest. Its potential benefit on minimizing myocardial ischemic injury has been explored, but clinical evidence has yet to confirm positive results in preclinical studies. Importantly, therapeutic hypothermia for myocardial infarction is unique in that it can be initiated prior to reperfusion, in contrast to its application for brain injury in resuscitated cardiac arrest patients. Recent advance in cooling technology allows more rapid cooling of the heart than ever and new clinical trials are designed to examine the efficacy of rapid therapeutic hypothermia for myocardial infarction. In this review, we summarize current knowledge regarding the effect of hypothermia on normal and ischemic hearts and discuss issues to be solved in order to realize its clinical application for treating acute myocardial infarction.
Collapse
Affiliation(s)
- Kelly P Yamada
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Taro Kariya
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tadao Aikawa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kiyotake Ishikawa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
9
|
Park J, Kwak JE, Cho YJ, Choi HJ, Choi H, Chae MS, Park CS, Choi JH, Hong SH. Therapeutic hypothermia after cardiac arrest during living-donor liver transplant surgery: A case report. Medicine (Baltimore) 2020; 99:e22513. [PMID: 33157915 PMCID: PMC7647609 DOI: 10.1097/md.0000000000022513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
RATIONALE Therapeutic hypothermia is an effective medical treatment for neurological recovery after cardiac arrest. Here, we describe a case of successful mild therapeutic hypothermia after cardiac arrest during living-donor liver transplantation. PATIENT CONCERNS A 54-year-old woman with alcoholic liver cirrhosis was admitted for living-donor liver transplantation. Cardiac arrest occurred during the anhepatic phase. After cardiopulmonary resuscitation, spontaneous circulation returned, but the bispectral index level remained below 10 until the end of surgery. DIAGNOSES Neurological injury caused by global cerebral hypoperfusion was suspected. INTERVENTIONS The patient was treated with mild therapeutic hypothermia for 24hours after resuscitation targeting a core body temperature of 34°C with surface cooling using ice bags. OUTCOMES The patient recovered consciousness about 22 hours after the event. However, she showed symptoms of delirium even when discharged. At the 3-month follow-up exam, she showed no specific neurological complications. The transplanted liver showed no problems with regeneration. LESSONS Mild therapeutic hypothermia may be safely adopted in cases of cardiac arrest in liver transplant patients and is beneficial for neurological recovery.
Collapse
Affiliation(s)
- Jaesik Park
- Department of Anesthesiology and Pain Medicine
| | - Ju Eun Kwak
- Department of Anesthesiology and Pain Medicine
| | | | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hoon Choi
- Department of Anesthesiology and Pain Medicine
| | | | | | | | | |
Collapse
|
10
|
Howard BT, Iaizzo PA. Induced functional modulations of isolated large mammalian hearts. Pflugers Arch 2019; 471:1095-1101. [PMID: 31123804 DOI: 10.1007/s00424-019-02277-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/25/2019] [Accepted: 04/16/2019] [Indexed: 11/25/2022]
Abstract
In this study we used Visible Heart® methodologies featuring cyclic temperature modulation of porcine hearts in order to establish characteristic temperature responses. This isolated and perfused model is a more predictable and modifiable analog for human heart preservation and isolates the response of the cardiac tissue. We comprehensively monitored isolated porcine hearts undergoing temperature change and demonstrated optimization of isolated cardiac function under mild hypothermia. We tracked metrics of cardiac function as continuous variables during temperature changes (~ 31 to 39 °C), eliciting a well-defined reduction in metabolic demand and in heart rate modulation. Optimization of function appeared to occur around 34.7 ± 0.9 °C (n = 13). Cardiac response was further investigated in the presence of active pacing in order to assess pacing capture and the heart's functional response without a means of regulating rate. Our results may have direct clinical implications for emerging heart preservation methods prior to transplantation, as well as benefits for investigators using isolated heart models for preclinical device testing. Clinically, this porcine model is a basis for finding new ways to extend the window of viability for transplantable organs, thereby restoring or improving graft function and potentially enhancing recipient outcomes.
Collapse
Affiliation(s)
- Brian T Howard
- Medtronic Inc., 8200 Coral Sea St NE, Mounds View, MN, 55112, USA
| | - Paul A Iaizzo
- Department of Surgery, University of Minnesota, 420 Delaware St. SE, B172 Mayo, MMC 195, Minneapolis, MN, 55455, USA.
| |
Collapse
|
11
|
Eskla KL, Porosk R, Reimets R, Visnapuu T, Vasar E, Hundahl CA, Luuk H. Hypothermia augments stress response in mammalian cells. Free Radic Biol Med 2018; 121:157-168. [PMID: 29704622 DOI: 10.1016/j.freeradbiomed.2018.04.571] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 04/16/2018] [Accepted: 04/22/2018] [Indexed: 12/22/2022]
Abstract
Mild hypothermia (32 °C) is routinely used in medical practice to alleviate hypoxic ischemic damage, however, the mechanisms that underlie its protective effects remain uncertain. Using a systems approach based on genome-wide expression screens, reporter assays and biochemical studies, we find that cellular hypothermia response is associated with the augmentation of major stress-inducible transcription factors Nrf2 and HIF1Α affecting the antioxidant system and hypoxia response pathways, respectively. At the same time, NF-κB, a transcription factor involved in the control of immune and inflammatory responses, was not induced by hypothermia. Furthermore, mild hypothermia did not trigger unfolded protein response. Lower temperatures (27 °C and 22 °C) did not activate Nrf2 and HIF1A pathways as efficiently as mild hypothermia. Current findings are discussed in the context of the thermodynamic hypothesis of therapeutic hypothermia. We argue that the therapeutic effects are likely to stem both from metabolic suppression (inhibitory component) and augmentation of stress tolerance (activating component). We argue that systems coping with cellular stressors are plausible targets of therapeutic hypothermia and deserve more attention in clinical hypothermia research.
Collapse
Affiliation(s)
- Kattri-Liis Eskla
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia.
| | - Rando Porosk
- Institute of Biomedicine and Translational Medicine, Department of Biochemistry, University of Tartu, Tartu, Estonia
| | - Riin Reimets
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia
| | - Tanel Visnapuu
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia
| | - Eero Vasar
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Christian Ansgar Hundahl
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia
| | - Hendrik Luuk
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
12
|
Bergan HA, Halvorsen PS, Espinoza A, Kerans V, Skulstad H, Fosse E, Bugge JF. Left Ventricle Function During Therapeutic Hypothermia with Beta 1-Adrenergic Receptor Blockade. Ther Hypothermia Temp Manag 2018; 8:156-164. [PMID: 29394143 DOI: 10.1089/ther.2017.0051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Therapeutic hypothermia is an established treatment in patients resuscitated from cardiac arrest. It is usually well-tolerated circulatory, but hypothermia negatively effects myocardial contraction and relaxation velocities and increases diastolic filling restrictions. A significant proportion of resuscitated patients are treated with long-acting beta-receptor blocking agents' prearrest, but the combined effects of hypothermia and beta-blockade on left ventricle (LV) function are not previously investigated. We hypothesized that beta1-adrenergic receptor blockade (esmolol infusion) exacerbates the negative effects of hypothermia on active myocardial motions, affecting both systolic and diastolic LV function. A pig (n = 10) study was performed to evaluate the myocardial effects of esmolol during hypothermia (33°C) and during normothermia, at spontaneous and pacing-increased heart rates (HRs). LV function was assessed by a LV pressure transducer, an epicardial ultrasonic transducer (wall thickness, wall thickening/thinning velocity) and an aortic ultrasonic flow-probe (stroke volume, cardiac output). The data were compared using a paired two-tailed Students t-test, and also analyzed using a linear mixed model to handle dependencies introduced by repeated measurements within each subject. The significance level was p ≤ 0.05. The effects of hypothermia and beta blockade were distinct and additive. Hypothermia reduced myocardial motion velocities and increased diastolic filling restrictions, but end-systolic wall thickness increased, and stroke volume and dP/dtmax (pumping function) were maintained. In contrast, esmolol predominantly affected systolic pumping function, by a negative inotropic effect. In combination, hypothermia and esmolol reduced myocardial velocities in systole and diastole by ∼40%, compared with normothermia without esmolol, inducing in combination both systolic and diastolic LV function impairment. The cardiac dysfunction deteriorated at increased HRs during hypothermia. Beta1-adrenergic receptor blockade (esmolol) exacerbates the negative effects of hypothermia on active myocardial contraction and relaxation. The combination of hypothermia with beta-blockade induces both systolic and diastolic LV function impairment.
Collapse
Affiliation(s)
- Harald A Bergan
- 1 Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital , Oslo, Norway .,2 Faculty of Medicine, Institute of Clinical Medicine, University of Oslo , Oslo, Norway
| | - Per S Halvorsen
- 3 The Intervention Centre, Rikshospitalet, Oslo University Hospital , Oslo, Norway
| | - Andreas Espinoza
- 1 Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital , Oslo, Norway
| | - Viesturs Kerans
- 1 Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital , Oslo, Norway .,3 The Intervention Centre, Rikshospitalet, Oslo University Hospital , Oslo, Norway
| | - Helge Skulstad
- 2 Faculty of Medicine, Institute of Clinical Medicine, University of Oslo , Oslo, Norway .,4 Department of Cardiology, Rikshospitalet, Oslo University Hospital , Oslo, Norway
| | - Erik Fosse
- 2 Faculty of Medicine, Institute of Clinical Medicine, University of Oslo , Oslo, Norway .,3 The Intervention Centre, Rikshospitalet, Oslo University Hospital , Oslo, Norway
| | - Jan F Bugge
- 1 Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital , Oslo, Norway .,2 Faculty of Medicine, Institute of Clinical Medicine, University of Oslo , Oslo, Norway
| |
Collapse
|
13
|
McGarvey M, Ali O, Iqbal MB, Ilsley C, Wong J, Di Mario C, Redwood S, Patterson T, Pennell DJ, Rogers P, Dalby M. A feasibility and safety study of intracoronary hemodilution during primary coronary angioplasty in order to reduce reperfusion injury in myocardial infarction. Catheter Cardiovasc Interv 2018. [PMID: 28636165 DOI: 10.1002/ccd.27136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES We designed a pilot study to evaluate safety and feasibility of an inexpensive and simple approach to intracoronary hemodilution during primary angioplasty (PPCI) to reduce reperfusion injury. INTRODUCTION Early revascularization in acute myocardial infarction decreases infarct size and improves outcomes. However, abrupt restoration of coronary flow results in myocardial reperfusion injury and increased final infarct size. Dilution of coronary blood during revascularization may help reduce this damage. If proved effective, such an approach would need to be simple and suitable for widespread adoption. METHODS Ten patients presenting with STEMI underwent intracoronary dilution with room temperature Hartmann's solution delivered through the guiding catheter during primary angioplasty (PPCI). Infusion of perfusate began prior to crossing the occluded artery with the guidewire, continuing until 10 min after completion of the balloon and stenting procedure. Infusion was briefly interrupted for contrast injection and pressure monitoring. The outcome measures were safety, including intracoronary temperature reduction and volume of intracoronary perfusate infused, and technical feasibility. RESULTS There were no significant symptomatic, hemodynamic, ECG ST/T segment or rhythm changes observed during perfusate administration. The median (interquartile range) volume of perfusate administered was 550 mL (350-725 mL) and the median intracoronary temperature reduction observed was 3.4°Celsius. Myocardial salvage was 0.54 (0.43-0.65). CONCLUSIONS Transcatheter intracoronary hemodilution with room temperature perfusate during PPCI is feasible and appears safe. Such a strategy is simple and inexpensive, with potential to be widely applied. Further mechanistic and subsequent outcome powered studies are required to evaluate whether this strategy can reduce reperfusion injury in STEMI.
Collapse
Affiliation(s)
- Michael McGarvey
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Omar Ali
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - M Bilal Iqbal
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Charles Ilsley
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Joyce Wong
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Carlo Di Mario
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Simon Redwood
- Department of Cardiology, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Tiffany Patterson
- Department of Cardiology, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Dudley J Pennell
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Paula Rogers
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | - Miles Dalby
- Department of Cardiology, NIHR Cardiovascular Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, and Imperial College London, United Kingdom
| | | |
Collapse
|
14
|
Zhao X, Gu T, Xiu Z, Shi E, Yu L. Mild Hypothermia May Offer Some Improvement to Patients with MODS after CPB Surgery. Braz J Cardiovasc Surg 2017; 31:246-251. [PMID: 27737408 PMCID: PMC5062708 DOI: 10.5935/1678-9741.20160048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 06/08/2016] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE: To summarize the effect of mild hypothermia on function of the organs in
patients with multiple organ dysfunction syndrome after cardiopulmonary
bypass surgery. METHODS: The patients were randomly divided into two groups, northermia group (n=71)
and hypothermia group (n=89). We immediately began cooling the hypothermia
group when test results showed multiple organ dysfunction syndrome,
meanwhile all patients of two groups were drawn blood to test blood gas,
liver and kidney function, blood coagulation function, and evaluated the
cardiac function using echocardiography from 12 to 36 hours. We compared the
difference of intra-aortic balloon pump, extracorporeal membrane oxygenation
rate and mortality within one month after intensive care unit admission. RESULTS: Among the 160 patients, 36 died, 10 (11.24%) patients were from the
hypothermia group and 26 (36.6%) from the northermia group
(P <0.05). In northermia group, 45 (63.38%) patients
used intra-aortic balloon pump and 4 (5.63%), extracorporeal membrane
oxygenation; in hypothermia group, 35 (39.32%) patients used intra-aortic
balloon pump and 2 (2.25%), extracorporeal membrane oxygenation(
P <0.05). The patients' heart rate decreased
significantly in the hypothermia group. The heart rate of hypothermia group
is significantly slower than the northermia group at the 36th
hour (P <0.05). But the mean arterial pressure of
hypothermia group is significantly higher than the northermia group at the
36th hour (P <0.05). In hypothermia
group, PO2, SvO2 and lactate were improved
significantly compared to pre-cooling (P <0.05), and
they were significantly better than the northermia group at the
36th hour (P <0.05%). Prothrombin time
and activated partial thromboplastin time have no significantly difference
between the two groups (P >0.05). But the platelet count
has significantly difference between the two groups at the 36th
hour (P <0.05). The aspartate transaminase, alanine
transaminase and creatinine were improved significantly in the hypothermia
group, and they were significantly better than the northermia group
(P <0.05). CONCLUSION: Mild hypothermia is feasible and safe for patients with multiple organ
dysfunction syndrome after cardiopulmonary bypass surgery.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Cardiac Surgery ICU, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Tianxiang Gu
- Department of Cardiac Surgery ICU, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Zongyi Xiu
- Department of Cardiac Surgery ICU, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Enyi Shi
- Department of Cardiac Surgery ICU, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Lei Yu
- Department of Cardiac Surgery ICU, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
15
|
Crombez T, Hachimi-Idrissi S. The influence of targeted temperature management on the pharmacokinetics of drugs administered during and after cardiac arrest: a systematic review. Acta Clin Belg 2017; 72:116-122. [PMID: 28220713 DOI: 10.1080/17843286.2017.1291782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Pharmacokinetic parameters of drugs are widely investigated under normothermic conditions and normal hemodynamic parameters. The European Resuscitation Council recommends the use of targeted temperature management (TTM) with a target temperature of 34 °C in cardiac arrest (CA) patients. The aim of this literature review is to investigate the influence of CA combined with TTM on the pharmacokinetics of drugs. Results of preclinical and clinical studies are compared with each other. Only the most important drugs, administered during CA in emergency setting, were studied. METHODS A literature search was conducted within PubMed and Google Scholar. The search terms included 'therapeutic hypothermia', 'TTM', 'drug metabolism', 'pharmacokinetics during hypothermia', 'cardiac arrest/etiology'. In Pubmed, MeSH-terms were also included: 'myocardial infarction/therapy', 'heart arrest/complications' and 'hypothermia'. To search for preclinical studies: the search terms 'pigs' and 'swine' were used. After the primary shift of relevant findings, further articles were found through references of these (snowballing method), as well as through related articles as suggested by the databases. RESULTS Due to the reduced cardiac output during TTM, most of the distribution volume ([Formula: see text]) of drugs included in this literature study is decreased. Only the [Formula: see text] of chlorzoxazone in CA rats and midazolam in non-CA patients are significantly increased during respectively deep and mild hypothermia. The renal, hepatic and biliary clearance of drugs administered during CA/TTM/hypothermia are decreased. DISCUSSION The combination of a decreased [Formula: see text] and a decrease in the metabolization/excretion of drugs during CA/TTM result in higher plasma concentrations compared to the plasma concentrations during CA without TTM.
Collapse
Affiliation(s)
- Tessa Crombez
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Said Hachimi-Idrissi
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Emergency Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
16
|
Otterspoor LC, van Nunen LX, Rosalina TT, Veer MV, Tuijl SV, Stijnen M, Rutten MCM, van de Vosse FN, Pijls NHJ. Intracoronary hypothermia for acute myocardial infarction in the isolated beating pig heart. Am J Transl Res 2017; 9:558-568. [PMID: 28337283 PMCID: PMC5340690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/06/2017] [Indexed: 06/06/2023]
Abstract
Hypothermia may attenuate reperfusion injury and thereby improve acute myocardial infarction therapy. Systemic cooling trials failed to reduce infarct size, perhaps because the target temperature was not reached fast enough. The use of selective intracoronary hypothermia combined with intracoronary temperature monitoring allows for titrating to target temperature and optimizing the cooling rate. We aimed to the test the feasibility of intracoronary cooling for controlled, selective myocardial hypothermia in an isolated beating pig heart. In five porcine hearts the left anterior descending artery (LAD) was occluded by an over-the-wire balloon (OTWB). After occlusion, saline at 22°C was infused through the OTWB lumen for 5 minutes into the infarct area at a rate of 30 ml/min. Thereafter the balloon was deflated but infusion continued with saline at 4°C for 5 minutes. Distal coronary temperature was continuously monitored by a pressure/temperature guidewire. Myocardial temperature at several locations in the infarct and control areas was recorded using needle thermistors. In the occlusion phase, coronary temperature decreased by 11.4°C (range 9.4-12.5°C). Myocardial temperature throughout the infarct area decreased by 5.1°C (range 1.8-8.1°C) within three minutes. During the reperfusion phase, coronary temperature decreased by 6.2°C (range 4.1-10.3°C) and myocardial temperature decreased by 4.5°C (range 1.5-7.4°C). Myocardial temperature outside the infarct area was not affected. In the isolated beating pig heart with acute occlusion of the LAD, we were able to rapidly "induce, maintain, and control" a stable intracoronary and myocardial target temperature of at least 4°C below body temperature without side effects and using standard PCI equipment, justifying further studies of this technique in humans.
Collapse
Affiliation(s)
- Luuk C Otterspoor
- Department of Cardiology, Catharina Hospital EindhovenEindhoven, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| | - Lokien X van Nunen
- Department of Cardiology, Catharina Hospital EindhovenEindhoven, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| | - Tilaï T Rosalina
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| | - Marcel van’t Veer
- Department of Cardiology, Catharina Hospital EindhovenEindhoven, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| | | | | | - Marcel CM Rutten
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| | - Frans N van de Vosse
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| | - Nico HJ Pijls
- Department of Cardiology, Catharina Hospital EindhovenEindhoven, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of TechnologyThe Netherlands
| |
Collapse
|
17
|
Grejs AM, Gjedsted J, Thygesen K, Lassen JF, Rasmussen BS, Jeppesen AN, Duez CHV, Søreide E, Kirkegaard H. The Extent of Myocardial Injury During Prolonged Targeted Temperature Management After Out-of-Hospital Cardiac Arrest. Am J Med 2017; 130:37-46. [PMID: 27477668 DOI: 10.1016/j.amjmed.2016.06.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 12/23/2022]
Abstract
AIM The aim of this study is to evaluate the extent of myocardial injury by cardiac biomarkers during prolonged targeted temperature management of 24 hours vs 48 hours after out-of-hospital cardiac arrest. METHODS This randomized Scandinavian multicenter study compares the extent of myocardial injury quantified by area under the curve (AUC) of cardiac biomarkers during prolonged targeted temperature management at 33°C ± 1°C of 24 hours and 48 hours, respectively. Through a period of 2.5 years, 161 comatose out-of-hospital cardiac arrest patients were randomized to targeted temperature management for 24 hours (n = 77) or 48 hours (n = 84). The AUC was calculated using both high-sensitivity cardiac troponin T (hs-cTnTAUC) and creatine kinase-myocardial band (CK-MBAUC) that were based upon measurements of these biomarkers every 6 hours upon admission until 96 hours after reaching target temperature. RESULTS The median hs-cTnTAUC of 33,827 ng/L/h (interquartile range [IQR] 11,366-117,690) of targeted temperature management at 24 hours did not differ significantly from that of 28,973 ng/L/h (IQR 10,656-163,655) at 48 hours. In contrast, the median CK-MBAUC of 1829 μg/L/h (IQR 800-6799) during targeted temperature management at 24 hours was significantly lower than that of 2428 μg/L/h (IQR 1163-10,906) within targeted temperature management at 48 hours, P <.05. CONCLUSION This study of comatose out-of-hospital cardiac arrest survivors showed no difference between the extents of myocardial injury estimated by hs-cTnTAUC of prolonged targeted temperature management of 48 hours vs 24 hours, although the CK-MBAUC was significantly higher during 48 hours vs 24 hours. Hence, it seems unlikely that the duration of targeted temperature management has a beneficial effect on the extent of myocardial injury after out-of-hospital cardiac arrest, and may even have a worsening effect.
Collapse
Affiliation(s)
- Anders Morten Grejs
- Research Center for Emergency Medicine, Aarhus University Hospital, Denmark; Department of Anesthesiology and Intensive Care Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark.
| | - Jakob Gjedsted
- Department of Anesthesiology and Intensive Care Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | | | - Jens Flensted Lassen
- Department of Cardiology, Rigshospitalet, University Hospital of Copenhagen, Denmark
| | - Bodil Steen Rasmussen
- Department of Anesthesiology and Intensive Care Medicine, Aalborg University, Denmark; Department of Clinical Medicine, Aalborg University, Denmark
| | - Anni Nørgaard Jeppesen
- Research Center for Emergency Medicine, Aarhus University Hospital, Denmark; Department of Anesthesiology and Intensive Care Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Christophe Henri Valdemar Duez
- Research Center for Emergency Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Eldar Søreide
- Department of Anesthesiology and Intensive Care, Stavanger University Hospital, Norway; Department of Clinical Medicine, University of Bergen, Norway
| | - Hans Kirkegaard
- Research Center for Emergency Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| |
Collapse
|
18
|
Bergan HA, Halvorsen PS, Skulstad H, Fosse E, Bugge JF. Does therapeutic hypothermia during extracorporeal cardiopulmonary resuscitation preserve cardiac function? J Transl Med 2016; 14:345. [PMID: 27998282 PMCID: PMC5175383 DOI: 10.1186/s12967-016-1099-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/29/2016] [Indexed: 01/16/2023] Open
Abstract
Background Extracorporeal cardiopulmonary resuscitation (E-CPR) is increasingly used as a rescue method in the management of cardiac arrest and provides the opportunity to rapidly induce therapeutic hypothermia. The survival after a cardiac arrest is related to post-arrest cardiac function, and the application of therapeutic hypothermia post-arrest is hypothesized to improve cardiac outcome. The present animal study compares normothermic and hypothermic E-CPR considering resuscitation success, post-arrest left ventricular function and magnitude of myocardial injury. Methods After a 15-min untreated ventricular fibrillation, the pigs (n = 20) were randomized to either normothermic (38 °C) or hypothermic (32–33 °C) E-CPR. Defibrillation terminated ventricular fibrillation after 5 min of E-CPR, and extracorporeal support continued for 2 h, followed by warming, weaning and a stabilization period. Magnetic resonance imaging and left ventricle pressure measurements were used to assess left ventricular function pre-arrest and 5 h post-arrest. Myocardial injury was estimated by serum concentrations of cardiac TroponinT and Aspartate transaminase (ASAT). Results E-CPR resuscitated all animals and the hypothermic strategy induced therapeutic hypothermia within minutes without impairment of the resuscitation success rate. All animals suffered a severe global systolic left ventricular dysfunction post-arrest with 50–70% reductions in stroke volume, ejection fraction, wall thickening, strain and mitral annular plane systolic excursion. Serum concentrations of cardiac TroponinT and ASAT increased considerably post-arrest. No significant differences were found between the two groups. Conclusions Two-hour therapeutic hypothermia during E-CPR offers an equal resuscitation success rate, but does not preserve the post-arrest cardiac function nor reduce the magnitude of myocardial injury, compared to normothermic E-CPR. Trial registration FOTS 4611/13 registered 25 October 2012 Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1099-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Harald A Bergan
- Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital, Oslo, Norway. .,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Per S Halvorsen
- The Intervention Centre, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Helge Skulstad
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Erik Fosse
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,The Intervention Centre, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Jan F Bugge
- Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
19
|
Kohlhauer M, Berdeaux A, Ghaleh B, Tissier R. Therapeutic hypothermia to protect the heart against acute myocardial infarction. Arch Cardiovasc Dis 2016; 109:716-722. [DOI: 10.1016/j.acvd.2016.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 10/20/2022]
|
20
|
Li X, Liu M, Sun R, Zeng Y, Chen S, Zhang P. Protective approaches against myocardial ischemia reperfusion injury. Exp Ther Med 2016; 12:3823-3829. [PMID: 28101167 PMCID: PMC5228114 DOI: 10.3892/etm.2016.3877] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/16/2016] [Indexed: 01/04/2023] Open
Abstract
Myocardial ischemia-reperfusion is the leading cause for the events of cardiovascular disease, and is considered as a major contributor to the morbidity and mortality associated with coronary occlusion. The myocardial damage caused by ischemia-reperfusion injury constitutes the primary pathological manifestation of coronary artery disease. It results from the interaction between the substances that accumulate during ischemia and those that are delivered on reperfusion. The level of this damage can range from a small insult resulting in limited myocardial damage to a large injury culminating in myocyte death. Importantly, major ischemia-reperfusion injury to the heart can result in permanent disability or death. Given the worldwide prevalence of coronary artery disease, developing a strategy to provide cardioprotection against ischemia-reperfusion-induced damage is of great importance. Currently, the treatment of reperfusion injury following ischemia is primarily supportive, since no specific target-oriented therapy has been validated thus far. Nevertheless, therapeutic approaches to protect against myocardial ischemia-reperfusion injury remain an active area of investigation given the detrimental effects of this phenomenon.
Collapse
Affiliation(s)
- Xianchi Li
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Min Liu
- Department of Cardiology, Xuzhou Clinical School of Xuzhou Medical College, Xuzhou, Jiangsu 221009, P.R. China
| | - Rongrong Sun
- Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, Jiangsu 221009, P.R. China
| | - Yi Zeng
- Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, Jiangsu 221009, P.R. China
| | - Shuang Chen
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Peiying Zhang
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
21
|
Onukwufor JO, Kibenge F, Stevens D, Kamunde C. Hypoxia-reoxygenation differentially alters the thermal sensitivity of complex I basal and maximal mitochondrial oxidative capacity. Comp Biochem Physiol A Mol Integr Physiol 2016; 201:87-94. [DOI: 10.1016/j.cbpa.2016.06.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/17/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
|
22
|
Zhang J, Xue X, Xu Y, Zhang Y, Li Z, Wang H. The transcriptome responses of cardiomyocyte exposed to hypothermia. Cryobiology 2016; 72:244-50. [DOI: 10.1016/j.cryobiol.2016.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 03/07/2016] [Accepted: 03/28/2016] [Indexed: 11/26/2022]
|
23
|
Kohlhauer M, Berdeaux A, Kerber RE, Micheau P, Ghaleh B, Tissier R. Liquid Ventilation for the Induction of Ultrafast Hypothermia in Resuscitation Sciences: A Review. Ther Hypothermia Temp Manag 2016; 6:63-70. [DOI: 10.1089/ther.2015.0024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Matthias Kohlhauer
- Inserm, Unité 955, Equipe 03, Créteil, France
- Université Paris Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France
- Université Paris Est, École Nationale Vétérinaire d'Alfort, Maisons-Alfort Cedex, France
| | - Alain Berdeaux
- Inserm, Unité 955, Equipe 03, Créteil, France
- Université Paris Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France
- Université Paris Est, École Nationale Vétérinaire d'Alfort, Maisons-Alfort Cedex, France
| | - Richard E. Kerber
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Philippe Micheau
- Department of Mechanical Engineering, Université de Sherbrooke, Sherbrooke, Canada
| | - Bijan Ghaleh
- Inserm, Unité 955, Equipe 03, Créteil, France
- Université Paris Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France
- Université Paris Est, École Nationale Vétérinaire d'Alfort, Maisons-Alfort Cedex, France
| | - Renaud Tissier
- Inserm, Unité 955, Equipe 03, Créteil, France
- Université Paris Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France
- Université Paris Est, École Nationale Vétérinaire d'Alfort, Maisons-Alfort Cedex, France
| |
Collapse
|
24
|
Abstract
Mild therapeutic hypothermia of 32-35°C improved neurologic outcomes in outside hospital cardiac arrest survivor. Furthermore, in experimental studies on infarcted model and pilot studies on conscious patients with acute myocardial infarction, therapeutic hypothermia successfully reduced infarct size and microvascular resistance. Therefore, mild therapeutic hypothermia has received an attention as a promising solution for reduction of infarction size after acute myocardial infarction which are not completely solved despite of optimal reperfusion therapy. Nevertheless, the results from randomized clinical trials failed to prove the cardioprotective effects of therapeutic hypothermia or showed beneficial effects only in limited subgroups. In this article, we reviewed rationale for therapeutic hypothermia and possible mechanisms from previous studies, effective methods for clinical application to the patients with acute myocardial infarction, lessons from current clinical trials and future directions.
Collapse
Affiliation(s)
- In Sook Kang
- Department of Internal Medicine, Green Hospital, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ikeno Fumiaki
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Wook Bum Pyun
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea.
| |
Collapse
|
25
|
Nadeau M, Sage M, Kohlhauer M, Vandamme J, Mousseau J, Robert R, Tissier R, Praud JP, Walti H, Micheau P. Thermal Dynamics in Newborn and Juvenile Models Cooled by Total Liquid Ventilation. IEEE Trans Biomed Eng 2015; 63:1483-91. [PMID: 26552070 DOI: 10.1109/tbme.2015.2496938] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Total liquid ventilation (TLV) consists in filling the lungs with a perfluorocarbon (PFC) and using a liquid ventilator to ensure a tidal volume of oxygenated, CO 2 -free and temperature-controlled PFC. Having a much higher thermal capacity than air, liquid PFCs assume that the filled lungs become an efficient heat exchanger with pulmonary circulation. OBJECTIVE The objective of the present study was the development and validation of a parametric lumped thermal model of a subject in TLV. METHODS The lungs were modeled as one compartment in which the control volume varied as a function of the tidal volume. The heat transfer in the body was modeled as seven parallel compartments representing organs and tissues. The thermal model of the lungs and body was validated with two groups of lambs of different ages and weights (newborn and juvenile) undergoing an ultrafast mild therapeutic hypothermia induction by TLV. RESULTS The model error on all animals yielded a small mean error of -0.1 ±0.4 (°)C for the femoral artery and 0.0 ±0.1 (°)C for the pulmonary artery. CONCLUSION The resulting experimental validation attests that the model provided an accurate estimation of the systemic arterial temperature and the venous return temperature. SIGNIFICANCE This comprehensive thermal model of the lungs and body has the advantage of closely modeling the rapid thermal dynamics in TLV. The model can explain how the time to achieve mild hypothermia between newborn and juvenile lambs remained similar despite of highly different physiological and ventilatory parameters. The strength of the model is its strong relationship with the physiological parameters of the subjects, which suggests its suitability for projection to humans.
Collapse
|
26
|
Herring MJ, Hale SL, Dai W, Oskui PM, Kloner RA. Hypothermia in the setting of experimental acute myocardial infarction: a comprehensive review. Ther Hypothermia Temp Manag 2015; 4:159-67. [PMID: 25271792 DOI: 10.1089/ther.2014.0016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A door-to-balloon time of less than 90 minutes is the gold standard for reperfusion therapy to treat acute myocardial infarction (MI). Because 30-day mortality remains ∼ 10%, new methods must be cultivated to limit myocardial injury. Therapeutic hypothermia has long been experimentally used to attenuate myocardial necrosis during MI with promising results, but the treatment has yet to gain popularity among most clinicians. Hypothermia, in the basic science setting, has been achieved using many techniques. In our review, we examine past and current methods of inducing hypothermia, benefits and setbacks of such methods, current and future clinical trials, and potential mechanisms.
Collapse
Affiliation(s)
- Michael J Herring
- 1 Heart Institute, Good Samaritan Hospital , Los Angeles, California
| | | | | | | | | |
Collapse
|
27
|
Nadeau M, Sage M, Kohlhauer M, Robert R, Vandamne J, Mousseau J, Tissier R, Praud JP, Walti H, Micheau P. Liquid ventilator for ultrafast hypothermia induction in juvenile lambs: Preliminary results. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2015:1695-1698. [PMID: 26736603 DOI: 10.1109/embc.2015.7318703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Total liquid ventilation (TLV) is an emerging mechanical ventilation technique. In this technique, the lungs are filled with liquid perfluorocarbons (PFC) and a liquid ventilator assures ventilation by periodically renewing a volume of oxygenated, CO2 freed and temperature controlled PFC. A huge difference between conventional mechanical ventilation and TLV relates to the fact that PFCs are about 1500 times denser than air. Thus, the PFCs filled lungs turn into an efficient heat exchanger with the circulating blood. One of the most appealing utilization of the lungs as a heat exchanger in TLV is for ultrafast induction of mild therapeutic hypothermia (MTH) for neuroprotection and cardioprotection after ischemia-reperfusion injuries. This study aimed to perform ultrafast MTH induction by TLV in animals up to 25 kg, then perform a fast post-hypothermic rewarming while maintaining proper ventilation. A thermal model of the lamb and liquid ventilator was developed to predict the dynamic and the control strategy to adopt for MTH induction. Two juvenile lambs were instrumented with temperature sensors in the femoral artery, pulmonary artery, oesophagus, right eardrum and rectum. After stabilization in conventional mechanical ventilation, TLV was initiated with ultrafast MTH induction, followed by posthypothermic rewarming. Preliminary results in the two juvenile lambs reveal that the liquid ventilator Inolivent-6.0 can induce MTH by TLV in less than 2.5 min for systemic arterial blood and in less than 10 min for venous return, esophagus and eardrum. Rectal temperature reached MTH in respectively 19.4 and 17.0 min for both lambs. Experimental results were consistent with the model predictions. Moreover, blood gas analysis exhibited that the gas exchange in the lungs was maintained adequately for the entire experiments.
Collapse
|
28
|
Tong G, Walker C, Bührer C, Berger F, Miera O, Schmitt KRL. Moderate hypothermia initiated during oxygen–glucose deprivation preserves HL-1 cardiomyocytes. Cryobiology 2015; 70:101-8. [DOI: 10.1016/j.cryobiol.2014.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 10/24/2022]
|
29
|
Champion S, Voicu S, Deye N. Conséquences cardiovasculaires de l’hypothermie. MEDECINE INTENSIVE REANIMATION 2015. [DOI: 10.1007/s13546-015-1054-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
Yan L, Kudej RK, Vatner DE, Vatner SF. Myocardial ischemic protection in natural mammalian hibernation. Basic Res Cardiol 2015; 110:9. [PMID: 25613166 DOI: 10.1007/s00395-015-0462-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/08/2014] [Accepted: 01/07/2015] [Indexed: 12/31/2022]
Abstract
Hibernating myocardium is an important clinical syndrome protecting the heart with chronic myocardial ischemia, named for its assumed resemblance to hibernating mammals in winter. However, the effects of myocardial ischemic protection have never been studied in true mammalian hibernation, which is a unique strategy for surviving extreme winter environmental stress. The goal of this investigation was to test the hypothesis that ischemic stress may also be protected in woodchucks as they hibernate in winter. Myocardial infarction was induced by coronary occlusion followed by reperfusion in naturally hibernating woodchucks in winter with and without hibernation and in summer, when not hibernating. The ischemic area at risk was similar among groups. Myocardial infarction was significantly less in woodchucks in winter, whether hibernating or not, compared with summer, and was similar to that resulting after ischemic preconditioning. Whereas several genes were up or downregulated in both hibernating woodchuck and with ischemic preconditioning, one mechanism was unique to hibernation, i.e., activation of cAMP-response element binding protein (CREB). When CREB was upregulated in summer, it induced protection similar to that observed in the woodchuck heart in winter. The cardioprotection in hibernation was also mediated by endothelial nitric oxide synthase, rather than inducible nitric oxide synthase. Thus, the hibernating woodchuck heart is a novel model to study cardioprotection for two major reasons: (1) powerful cardioprotection occurs naturally in winter months in the absence of any preconditioning stimuli, and (2) it resembles ischemic preconditioning, but with novel mechanisms, making this model potentially useful for clinical translation.
Collapse
Affiliation(s)
- Lin Yan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | | | | | | |
Collapse
|
31
|
Nadeau M, Micheau P, Robert R, Avoine O, Tissier R, Germim PS, Vandamme J, Praud JP, Walti H. Core Body Temperature Control by Total Liquid Ventilation Using a Virtual Lung Temperature Sensor. IEEE Trans Biomed Eng 2014; 61:2859-68. [DOI: 10.1109/tbme.2014.2332293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Chenoune M, De Rochefort L, Bruneval P, Lidouren F, Kohlhauer M, Seemann A, Ghaleh B, Korn M, Dubuisson RM, Ben Yahmed A, Maître X, Isabey D, Ricard JD, Kerber RE, Darrasse L, Berdeaux A, Tissier R. Evaluation of lung recovery after static administration of three different perfluorocarbons in pigs. BMC Pharmacol Toxicol 2014; 15:53. [PMID: 25253660 PMCID: PMC4177717 DOI: 10.1186/2050-6511-15-53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 09/18/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The respiratory properties of perfluorocarbons (PFC) have been widely studied for liquid ventilation in humans and animals. Several PFC were tested but their tolerance may depend on the species. Here, the effects of a single administration of liquid PFC into pig lungs were assessed and compared. Three different PFC having distinct evaporative and spreading coefficient properties were evaluated (Perfluorooctyl bromide [PFOB], perfluorodecalin [PFD] and perfluoro-N-octane [PFOC]). METHODS Pigs were anesthetized and submitted to mechanical ventilation. They randomly received an intra-tracheal administration of 15 ml/kg of either PFOB, PFD or PFOC with 12 h of mechanical ventilation before awakening and weaning from ventilation. A Control group was submitted to mechanical ventilation with no PFC administration. All animals were followed during 4 days after the initial PFC administration to investigate gas exchanges and clinical recovery. They were ultimately euthanized for histological analyses and assessment of PFC residual concentrations within the lungs using dual nuclei fluorine and hydrogen Magnetic Resonance Imaging (MRI). Sixteen animals were included (4/group). RESULTS In the PFD group, animals tended to be hypoxemic after awakening. In PFOB and PFOC groups, blood gases were not significantly different from the Control group after awakening. The poor tolerance of PFD was likely related to a large amount of residual PFC, as observed using MRI in all lung samples (≈10% of lung volume). This percentage was lower in the PFOB group (≈1%) but remained significantly greater than in the Control group. In the PFOC group, the percentage of residual PFC was not significantly different from that of the Control group (≈0.1%). Histologically, the most striking feature was an alveolar infiltration with foam macrophages, especially in the groups treated by PFD or PFOB. CONCLUSIONS Of the three tested perfluorocarbons, PFOC offered the best tolerance in terms of lung function, gas exchanges and residuum in the lung. PFOC was rapidly cleared from the lungs and virtually disappeared after 4 days whereas PFOB persisted at significant levels and led to foam macrophage infiltration. PFOC could be relevant for short term total liquid ventilation with a rapid weaning.
Collapse
|
33
|
Demirgan S, Erkalp K, Sevdi MS, Aydogmus MT, Kutbay N, Firincioglu A, Ozalp A, Alagol A. Cardiac condition during cooling and rewarming periods of therapeutic hypothermia after cardiopulmonary resuscitation. BMC Anesthesiol 2014; 14:78. [PMID: 25258591 PMCID: PMC4174499 DOI: 10.1186/1471-2253-14-78] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 09/11/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Hypothermia has been used in cardiac surgery for many years for neuroprotection. Mild hypothermia (MH) [body temperature (BT) kept at 32-35°C] has been shown to reduce both mortality and poor neurological outcome in patients after cardiopulmonary resuscitation (CPR). This study investigated whether patients who were expected to benefit neurologically from therapeutic hypothermia (TH) also had improved cardiac function. METHODS The study included 30 patients who developed in-hospital cardiac arrest between September 17, 2012, and September 20, 2013, and had return of spontaneous circulation (ROSC) following successful CPR. Patient BTs were cooled to 33°C using intravascular heat change. Basal BT, systolic artery pressure (SAP), diastolic artery pressure (DAP), mean arterial pressure (MAP), heart rate, central venous pressure, cardiac output (CO), cardiac index (CI), global end-diastolic volume index (GEDI), extravascular lung water index (ELWI), and systemic vascular resistance index (SVRI) were measured at 36°C, 35°C, 34°C and 33°C during cooling. BT was held at 33°C for 24 hours prior to rewarming. Rewarming was conducted 0.25°C/h. During rewarming, measurements were repeated at 33°C, 34°C, 35°C and 36°C. A final measurement was performed once patients spontaneously returned to basal BT. We compared cooling and rewarming cardiac measurements at the same BTs. RESULTS SAP values during rewarming (34°C, 35°C and 36°C) were lower than during cooling (P < 0.05). DAP values during rewarming (basal temperature, 34°C, 35°C and 36°C) were lower than during cooling. MAP values during rewarming (34°C, 35°C and 36°C) were lower than during cooling (P < 0.05). CO and CI values were higher during rewarming than during cooling. GEDI and ELWI did not differ during cooling and rewarming. SVRI values during rewarming (34°C, 35°C, 36°C and basal temperature) were lower than during cooling (P < 0.05). CONCLUSIONS To our knowledge, this is the first study comparing cardiac function at the same BTs during cooling and rewarming. In patients experiencing ROSC following CPR, TH may improve cardiac function and promote favorable neurological outcomes.
Collapse
Affiliation(s)
- Serdar Demirgan
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - Kerem Erkalp
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - M Salih Sevdi
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - Meltem Turkay Aydogmus
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - Numan Kutbay
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - Aydin Firincioglu
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - Ali Ozalp
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| | - Aysin Alagol
- Department of Anesthesiology and Reanimation, Bagcilar Educational and Training Hospital, Şenlikköy Mah, İncir Sokak, No:1/3, Sarı Konaklar Sitesi, B-Blok, Daire:6, Florya/ Bakırköy, Istanbul, Turkey
| |
Collapse
|
34
|
Longnus SL, Mathys V, Dornbierer M, Dick F, Carrel TP, Tevaearai HT. Heart transplantation with donation after circulatory determination of death. Nat Rev Cardiol 2014; 11:354-63. [DOI: 10.1038/nrcardio.2014.45] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Chen CW, Puvanesarajah V, Lo SFL, Cheng TJ, Cheng CY, Lim M, Lin HJ. Selective cerebral hypothermia induced via hypothermic retrograde jugular vein saline flush in a porcine model. Neurol Res 2014; 36:897-902. [PMID: 24725291 DOI: 10.1179/1743132814y.0000000374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Multiple methods of selective brain cooling have been used to prevent cerebral ischemia secondary to trauma and pathological or iatrogenic cerebral blood flow restriction. In this study, we tested the efficacy of hypothermic retrograde jugular vein flush (HRJVF) in eliciting selective brain hypothermia in a porcine model. METHODS Twelve swine were divided into two groups: retrograde jugular vein infusion (RJVI) with cold saline (4°C RJVI, n = 6) and with room temperature saline (24°C RJVI, n = 6). For 90 minutes, the following parameters were measured: brain parenchymal temperature, rectal temperature, intracranial pressure (ICP), mean arterial pressure, and heart rate (HR). RESULTS Swine receiving 4°C RJVI experienced a drop in mean brain parenchymal temperature of 1·1 ± 0·1°C, compared to 0·1 ± 0·1°C in swine receiving 24°C RJVI. At 90 minutes, mean brain parenchymal temperature in the 4°C RJVI treatment group was 35·5 ± 0·2°C, as compared to 37·1 ± 0·2°C in the 24°C RJVI treatment group (P < 0·001). In the 4°C RJVI group, the brain-systemic temperature gradient peaked 10 minutes after initiation of cooling and remained significantly different when comparing the two experimental groups (P < 0·001) throughout the duration of the 90 minutes experiment. Of note, ICP, mean arterial pressure, and HR remained constant without any significant changes or differences between treatment groups. DISCUSSION These results suggest that HRJVF is an effective method for selective brain hypothermia in a large animal model. Clinical application may prove effective in delaying neural ischemia.
Collapse
|
36
|
Testori C, Sterz F, Delle-Karth G, Malzer R, Holzer M, Stratil P, Stöckl M, Weiser C, van Tulder R, Gangl C, Sebald D, Zajicek A, Buchinger A, Lang I. Strategic target temperature management in myocardial infarction--a feasibility trial. Heart 2013; 99:1663-7. [PMID: 24064228 PMCID: PMC3812852 DOI: 10.1136/heartjnl-2013-304624] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE The purpose of this study was to demonstrate the feasibility of a combined cooling strategy started out of hospital as an adjunctive to percutaneous coronary intervention (PCI) in the treatment of ST-elevation acute coronary syndrome (STE-ACS). DESIGN Non-randomised, single-centre feasibility trial. SETTING Department of emergency medicine of a tertiary-care facility, Medical University of Vienna, Vienna, Austria. In cooperation with the Municipal ambulance service of the city of Vienna. PATIENTS Consecutive patients with STE-ACS presenting to the emergency medical service within 6 h after symptom onset. INTERVENTIONS Cooling was initiated with surface cooling pads in the out-of-hospital setting, followed by the administration of 1000-2000 mL of cold saline at hospital arrival and completed by endovascular cooling in the catheterisation laboratory. MAIN OUTCOME MEASURES Feasibility of lowering core temperature below 35.0°C prior to immediately performed revascularisation. Safety and tolerability of the cooling procedure. RESULTS In enrolled 19 patients (one woman, median age 51 years (IQR 45-59)), symptom onset to first medical contact (FMC) was 45 min (IQR 31-85). A core temperature below 35.0°C at reperfusion of the culprit lesion was achieved in 11 patients (78%) within 100 min (IQR 90-111) after FMC without any cooling-related serious adverse event. Temperature could be lowered from baseline 36.4°C (IQR 36.2-36.5°C) to 34.4°C (IQR 34.1-35.0°C) at the time of reperfusion. CONCLUSIONS With limitations an immediate out-of-hospital therapeutic hypothermia strategy was feasible and safe in patients with STE-ACS undergoing primary PCI. CLINICAL TRIAL REGISTRATION http://www.clinicaltrials.gov/ct2/show/NCT01864343; clinical trials unique identifier: NCT01864343.
Collapse
Affiliation(s)
- Christoph Testori
- Department of Emergency Medicine, Medical University of Vienna, , Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Minasian SM, Galagudza MM, Dmitriev YV, Kurapeev DI, Vlasov TD. Myocardial protection against global ischemia with Krebs-Henseleit buffer-based cardioplegic solution. J Cardiothorac Surg 2013; 8:60. [PMID: 23547937 PMCID: PMC3654955 DOI: 10.1186/1749-8090-8-60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 03/25/2013] [Indexed: 11/23/2022] Open
Abstract
Background The Krebs-Henseleit buffer is the best perfusion solution for isolated mammalian hearts. We hypothesized that a Krebs-Henseleit buffer-based cardioplegic solution might provide better myocardial protection than well-known crystalloid cardioplegic solutions because of its optimal electrolyte and glucose levels, presence of buffer systems, and mild hyperosmolarity. Methods Isolated Langendorff-perfused rat hearts were subjected to either global ischemia without cardioplegia (controls) or cardioplegic arrest for either 60 or 180 min, followed by 120 min of reperfusion. The modified Krebs-Henseleit buffer-based cardioplegic solution (mKHB) and St. Thomas’ Hospital solution No. 2 (STH2) were studied. During global ischemia, the temperatures of the heart and the cardioplegic solutions were maintained at either 37°C (60 min of ischemia) or 22°C (moderate hypothermia, 180 min of ischemia). Hemodynamic parameters were registered throughout the experiments. The infarct size was determined through histochemical examination. Results Cardioplegia with the mKHB solution at moderate hypothermia resulted in a minimal infarct size (5 ± 3%) compared to that in the controls and STH2 solution (35 ± 7% and 19 ± 9%, respectively; P < 0.001, for both groups vs. the mKHB group). In contrast to the control and STH2-treated hearts, no ischemic contracture was registered in the mKHB group during the 180-min global ischemia. At normothermia, the infarct sizes were 4 ± 3%, 72 ± 6%, and 70 ± 12% in the mKHB, controls, and STH2 groups, respectively (P < 0.0001). In addition, cardioplegia with mKHB at normothermia prevented ischemic contracture and improved the postischemic functional recovery of the left ventricle (P < 0.001, vs. STH2). Conclusions The data suggest that the Krebs-Henseleit buffer-based cardioplegic might be superior to the standard crystalloid solution (STH2).
Collapse
Affiliation(s)
- Sarkis M Minasian
- Institute of Experimental Medicine, V, A, Almazov Federal Heart, Blood and Endocrinology Centre, Saint Petersburg, Russia
| | | | | | | | | |
Collapse
|
38
|
Nadeau M, Micheau P, Robert R, Avoine O, Tissier R, Germim PS, Walti H. Control of rapid hypothermia induction by total liquid ventilation: preliminary results. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2013:3757-3760. [PMID: 24110548 DOI: 10.1109/embc.2013.6610361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mild therapeutic hypothermia (MTH) consists in cooling the body temperature of a patient to between 32 and 34 °C. This technique helps to preserve tissues and neurological functions in multi-organ failure by preventing ischemic injury. Total liquid ventilation (TLV) ensures gas exchange in the lungs with a liquid, typically perfluorocarbon (PFC). A liquid ventilator is responsible for ensuring cyclic renewal of tidal volume of oxygenated and temperature-controlled PFC. Hence, TLV using the lung as a heat exchanger and PFC as a heat carrier allows ultra fast cooling of the whole body which can help improve outcome after ischemic injuries. The present study was aimed to evaluate the control performance and safety of automated ultrarapid MTH induction by TLV. Experimentation was conducted using the Inolivent-5.0 liquid ventilator equipped with a PFC treatment unit that allows PFC cooling and heating from the flow of energy carrier water inside a double wall installed on an oxygenator. A water circulating bath is used to manage water temperature. A feedback controller was developed to modulate inspired PFC temperature and control body temperature. Such a controller is important since, with MTH induction, heart temperature should not reach 28 °C because of a high risk of fibrillation. The in vivo experimental protocol was conducted on a male newborn lamb of 4.7 kg which, after anesthetization, was submitted to conventional gas ventilation and instrumented with temperature sensors at the femoral artery, oesophagus, right ear drum and rectum. After stabilization, TLV was initiated with fast automated MTH induction to 33.5 °C until stabilization of all temperatures. MTH could be reached safely in 3 minutes at the femoral artery, in 3.6 minutes at the esophagus, in 7.7 minutes at the eardrum and in 15 minutes at the rectum. All temperatures were stable at 33.5 ± 0.5 °C within 15 minutes. The present results reveal that ultra-fast MTH induction by TLV with Inolivent-5.0 is safe for the heart while maintaining esophageal and arterial temperature over 32.6 °C.
Collapse
|
39
|
Stadelmann M, Dornbierer M, Clément D, Gahl B, Dick F, Carrel TP, Tevaearai HT, Longnus S. Mild hypothermia during global cardiac ischemia opens a window of opportunity to develop heart donation after cardiac death. Transpl Int 2012. [DOI: 10.1111/tri.12022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Monika Dornbierer
- Department of Cardiovascular Surgery; Inselspital; Berne University Hospital and University of Berne; Switzerland
| | | | - Brigitta Gahl
- Department of Cardiovascular Surgery; Inselspital; Berne University Hospital and University of Berne; Switzerland
| | - Florian Dick
- Department of Cardiovascular Surgery; Inselspital; Berne University Hospital and University of Berne; Switzerland
| | - Thierry P. Carrel
- Department of Cardiovascular Surgery; Inselspital; Berne University Hospital and University of Berne; Switzerland
| | - Hendrik T. Tevaearai
- Department of Cardiovascular Surgery; Inselspital; Berne University Hospital and University of Berne; Switzerland
| | - Sarah Longnus
- Department of Cardiovascular Surgery; Inselspital; Berne University Hospital and University of Berne; Switzerland
| |
Collapse
|
40
|
Schwartz BG, Kloner RA, Thomas JL, Bui Q, Mayeda GS, Burstein S, Hale SL, Economides C, French WJ. Therapeutic hypothermia for acute myocardial infarction and cardiac arrest. Am J Cardiol 2012; 110:461-6. [PMID: 22541421 DOI: 10.1016/j.amjcard.2012.03.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/17/2012] [Accepted: 03/17/2012] [Indexed: 10/28/2022]
Abstract
This report focuses on cardioprotection and describes the advantages and disadvantages of various methods of inducing therapeutic hypothermia (TH) with regard to neuroprotection and cardioprotection for patients with cardiac arrest and ST-segment elevation myocardial infarction (STEMI). TH is recommended in cardiac arrest guidelines. For patients resuscitated after out-of-hospital cardiac arrest, improvements in survival and neurologic outcomes were observed with relatively slow induction of TH. More rapid induction of TH in patients with cardiac arrest might have a mild to modest incremental impact on neurologic outcomes. TH drastically reduces infarct size in animal models, but achievement of target temperature before reperfusion is essential. Rapid initiation of TH in patients with STEMI is challenging but attainable, and marked infarct size reductions are possible. To induce TH, a variety of devices have recently been developed that require additional study. Of particular interest is transcoronary induction of TH using a catheter or wire lumen, which enables hypothermic reperfusion in the absence of total-body hypothermia. At present, the main methods of inducing and maintaining TH are surface cooling, endovascular heat-exchange catheters, and intravenous infusion of cold fluids. Surface cooling or endovascular catheters may be sufficient for induction of TH in patients resuscitated after out-of-hospital cardiac arrest. For patients with STEMI, intravenous infusion of cold fluids achieves target temperature very rapidly but might worsen left ventricular function. More widespread use of TH would improve survival and quality of life for patients with out-of-hospital cardiac arrest; larger studies with more rapid induction of TH are needed in the STEMI population.
Collapse
|
41
|
Tissier R, Chenoune M, Pons S, Zini R, Darbera L, Lidouren F, Ghaleh B, Berdeaux A, Morin D. Mild hypothermia reduces per-ischemic reactive oxygen species production and preserves mitochondrial respiratory complexes. Resuscitation 2012; 84:249-55. [PMID: 22796545 DOI: 10.1016/j.resuscitation.2012.06.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/21/2012] [Accepted: 06/23/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND Mitochondrial dysfunction is critical following ischemic disorders. Our goal was to determine whether mild hypothermia could limit this dysfunction through per-ischemic inhibition of reactive oxygen species (ROS) generation. METHODS First, ROS production was evaluated during simulated ischemia in an vitro model of isolated rat cardiomyocytes at hypothermic (32°C) vs. normothermic (38°C) temperatures. Second, we deciphered the direct effect of hypothermia on mitochondrial respiration and ROS production in oxygenated mitochondria isolated from rabbit hearts. Third, we investigated these parameters in cardiac mitochondria extracted after 30-min of coronary artery occlusion (CAO) under normothermic conditions (CAO-N) or with hypothermia induced by liquid ventilation (CAO-H; target temperature: 32°C). RESULTS In isolated rat cardiomyocytes, per-ischemic ROS generation was dramatically decreased at 32 vs. 38°C (e.g., -55±8% after 140min of hypoxia). In oxygenated mitochondria isolated from intact rabbit hearts, hypothermia also improved respiratory control ratio (+22±3%) and reduced H2O2 production (-41±1%). Decreased oxidative stress was further observed in rabbit hearts submitted to hypothermic vs. normothermic ischemia (CAO-H vs. CAO-N), using thiobarbituric acid-reactive substances as a marker. This was accompanied by a preservation of the respiratory control ratio as well as the activity of complexes I, II and III in cardiac mitochondria. CONCLUSION The cardioprotective effect of mild hypothermia involves a direct effect on per-ischemic ROS generation and results in preservation of mitochondrial function. This might explain why the benefit afforded by hypothermia during regional myocardial ischemia depends on how fast it is instituted during the ischemic process.
Collapse
|
42
|
|
43
|
Gerczuk PZ, Kloner RA. An update on cardioprotection: a review of the latest adjunctive therapies to limit myocardial infarction size in clinical trials. J Am Coll Cardiol 2012; 59:969-78. [PMID: 22402067 DOI: 10.1016/j.jacc.2011.07.054] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 06/13/2011] [Accepted: 07/19/2011] [Indexed: 12/13/2022]
Abstract
Acute myocardial infarction (AMI) with subsequent left ventricular dysfunction and heart failure continues to be a major cause of morbidity and mortality in the Western world. Rapid advances in the treatment of AMI, mainly through timely reperfusion, have substantially improved outcomes in patients presenting with acute coronary syndrome and particularly ST-segment elevation myocardial infarction. A vast amount of research, both translational and clinical, has been published on various pharmacological and interventional techniques to prevent myocardial cell death during the time of ischemia and subsequent reperfusion. Several methods of cardioprotection have shown the ability to limit myocardial infarction size in clinical trials. Examples of interventional techniques that have proven beneficial are ischemic post-conditioning and remote ischemic per-conditioning, both of which can reduce infarction size. Lowering core body temperature with cold saline infusion and cooling catheters have also been shown to be effective in certain circumstances. The most promising pharmaceutical cardioprotective agents at this time appear to be adenosine, atrial natriuretic peptide, and cyclosporine, with other potentially effective medications in the pipeline. Additional pre-clinical and clinical research is needed to further investigate newer cardioprotective strategies to continue the current trend of improving outcomes following AMI.
Collapse
Affiliation(s)
- Paul Z Gerczuk
- Heart Institute, Good Samaritan Hospital, Wilshire Boulevard, Los Angeles, CA 90017, USA
| | | |
Collapse
|
44
|
Dae MW. Hypothermia and percutaneous coronary intervention during acute myocardial infarction. Interv Cardiol 2012. [DOI: 10.2217/ica.12.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
45
|
Vander Heide R. Clinically useful cardioprotection: ischemic preconditioning then and now. J Cardiovasc Pharmacol Ther 2012; 16:251-4. [PMID: 21821524 DOI: 10.1177/1074248411407070] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ischemic preconditioning (IP) is the most effective, reproducible form of protection against myocardial cell death yet described. The mechanism of classic IP has not been identified, but recent investigations have focused on the mitochondrial permeability transition pore (mPTP). Similarly, the mechanism of the "second window of protection" (SWOP) is not known but thought to involve increased expression of important gene products. Currently, IP in the clinical arena is limited to cardiac surgery, planned angioplasty, and organ preservation protocols. To move preconditioning into a broader clinical arena will require resolution of important fundamental yet stubborn problems involving both basic and clinical science. Important unresolved issues include the mechanisms involved in the transition from reversible to irreversible injury, the amount of potential salvageable myocardium present at the onset of reperfusion, the identity and signaling of the mPTP, the optimization of protocols, the identity of end effectors (SWOP), and the identification of the best experimental model systems. From a clinical standpoint, important issues include the influence of comorbidities on cardioprotection, identification of appropriate animal models, the lack of a biologic marker of the cardioprotective state, the influence of coexistent therapeutic drugs, potential toxicity of pharmacologic mimics, and the window of opportunity for significant protection. Ischemic preconditioning has yielded promising results in other organs including the brain as well as tissue preservation for certain surgical procedures that will require definition of the underlying mechanism(s) to be fully exploited clinically. Over the past 25 years, the scientific community has learned much regarding the biology and potential mechanisms of IP and the concept has been expanded to many other organ systems in many other clinically relevant scenarios. To realize the full clinical potential will require continued investigation into the mechanism.
Collapse
Affiliation(s)
- Richard Vander Heide
- Pathology Department, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
46
|
Cohen MV, Downey JM. Is it time to translate ischemic preconditioning's mechanism of cardioprotection into clinical practice? J Cardiovasc Pharmacol Ther 2012; 16:273-80. [PMID: 21821528 DOI: 10.1177/1074248411407071] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
After three decades of intense research on cardioprotection, we still do not have an approved intervention for limiting infarct size in the patient with acute myocardial infarction (AMI) aside from reperfusion therapy. Yet approximately 25% of patients with AMI that are reperfused are still at risk for heart failure because of excessive muscle necrosis. This article will try to make the case that ischemic preconditioning (IPC), still the most potent anti-infarct intervention ever described, is ready for serious clinical testing now. Over the past 25 years, IPC's mechanism has been largely elucidated and targets a reperfusion injury. Ischemic preconditioning was never considered an intervention for AMI because of its need for pretreatment. However, knowledge of IPC's mechanism has revealed a large number of drugs and interventions that will activate IPC's signaling pathway at the time of reperfusion. Several small clinical trials suggest that they can be quite effective, but so far industry seems to have little interest in developing them. So, while basic scientists are in a continuous cycle of discovery and publication for new and novel cardioprotectants, there has been little effort devoted to translating these interventions into clinical practice. We believe that there are suitable IPC-based interventions that are ready for clinical testing today and the time has come for large-scale clinical trials.
Collapse
Affiliation(s)
- Michael V Cohen
- Department of Physiology, University of South Alabama, College of Medicine, Mobile, AL 36688, USA.
| | | |
Collapse
|
47
|
Assaly R, de Tassigny AD, Paradis S, Jacquin S, Berdeaux A, Morin D. Oxidative stress, mitochondrial permeability transition pore opening and cell death during hypoxia-reoxygenation in adult cardiomyocytes. Eur J Pharmacol 2011; 675:6-14. [PMID: 22173126 DOI: 10.1016/j.ejphar.2011.11.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/21/2011] [Accepted: 11/27/2011] [Indexed: 12/21/2022]
Abstract
Reactive oxygen species production is necessary to induce cell death following hypoxia/reoxygenation but the effect of reactive oxygen species produced during hypoxia on mitochondrial permeability transition pore (mPTP) opening and cell death is not established. Here we designed a model of hypoxia/reoxygenation in isolated cardiomyocytes measuring simultaneously reactive oxygen species production, mPTP opening and cell death in order (i) to establish a causal relationship between them, and (ii) to investigate the roles of various reactive oxygen species in mPTP opening. The percentage of cardiomyocytes exhibiting mPTP opening during reoxygenation increased with the duration of hypoxia. Antioxidants increased the time to mPTP opening when present during hypoxia but not at reoxygenation. This was associated with a drop in hydroxyl radical and hydrogen peroxide during hypoxia and the first minutes of reoxygenation. The increase in time to mPTP opening was accompanied by an improvement in cell viability reflected by maintenance of superoxide production at reoxygenation. Cyclosporin A delayed both the time to mPTP opening and cell death despite maintenance of reactive oxygen species production during hypoxia. These findings demonstrate that reactive oxygen species production precedes mPTP opening and that reactive oxygen species produced during hypoxia, particularly hydroxyl radicals and hydrogen peroxide, are necessary to induce mPTP opening which depends on hypoxia duration.
Collapse
Affiliation(s)
- Rana Assaly
- INSERM U955 équipe 03, F-94010, Créteil, France
| | | | | | | | | | | |
Collapse
|
48
|
Tissier R, Ghaleh B, Cohen MV, Downey JM, Berdeaux A. Myocardial protection with mild hypothermia. Cardiovasc Res 2011; 94:217-25. [PMID: 22131353 DOI: 10.1093/cvr/cvr315] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mild hypothermia, 32-35° C, is very potent at reducing myocardial infarct size in rabbits, dogs, sheep, pigs, and rats. The benefit is directly related to reduction in normothermic ischaemic time, supporting the relevance of early and rapid cooling. The cardioprotective effect of mild hypothermia is not limited to its recognized reduction of infarct size, but also results in conservation of post-ischaemic contractile function, prevention of no-reflow or microvascular obstruction, and ultimately attenuation of left ventricular remodelling. The mechanism of the anti-infarct effect does not appear to be related to diminished energy utilization and metabolic preservation, but rather to survival signalling that involves either the extracellular signal-regulated kinases and/or the Akt/phosphoinositide 3-kinase/mammalian target of rapamycin pathways. Initial clinical trials of hypothermia in patients with ST-segment elevation myocardial infarction were disappointing, probably because cooling was too slow to shorten normothermic ischaemic time appreciably. New approaches to more rapid cooling have recently been described and may soon be available for clinical use. Alternatively, it may be possible to pharmacologically mimic the protection provided by cooling soon after the onset of ischaemia with an activator of mild hypothermia signalling, e.g. extracellular signal-regulated kinase activator, that could be given by emergency medical personnel. Finally, the protection afforded by cooling can be added to that of pre- and post-conditioning because their mechanisms differ. Thus, myocardial salvage might be greatly increased by rapidly cooling patients as soon as possible and then giving a pharmacological post-conditioning agent immediately prior to reperfusion.
Collapse
|
49
|
Erlinge D. A Review of Mild Hypothermia as an Adjunctive Treatment for ST-Elevation Myocardial Infarction. Ther Hypothermia Temp Manag 2011; 1:129-41. [DOI: 10.1089/ther.2011.0008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- David Erlinge
- Department of Cardiology, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
50
|
Hypothermia down-regulates the LPS-induced norepinephrine (NE) release in ischaemic human heart cells. Brain Res Bull 2011; 87:67-73. [PMID: 21963948 DOI: 10.1016/j.brainresbull.2011.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/17/2011] [Accepted: 09/19/2011] [Indexed: 11/21/2022]
Abstract
Hypothermia has been widely acknowledged as the fundamental component of myocardial protection during cardiac operations. In this work, we studied in human atrial tissue the effect of the common hypothermic protection used in cardiac surgery, and we assessed this effect by comparing catecholamine release among normoxic, ischaemic, and inflammatory conditions. Our results provide the first evidence that lipopolysaccharide treatment results in an extremely dramatic and significant increase in the resting norepinephrine release under ischaemic conditions that can be normalised by hypothermia. These findings demonstrate that inflammatory conditions increase the temperature sensitivity of the norepinephrine transporter in human cardiac tissue. When the possible pharmacological interventions are taken into consideration, the results presented here provide new insight into the protection against ischaemia/reperfusion injury during cardiac surgery.
Collapse
|