1
|
Alotaibi K, Arulkumaran N, Dyson A, Singer M. Therapeutic strategies to ameliorate mitochondrial oxidative stress in ischaemia-reperfusion injury: A narrative review. Clin Sci (Lond) 2025; 139:CS20242074. [PMID: 39899361 DOI: 10.1042/cs20242074] [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: 09/12/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025]
Abstract
Mitochondrial reactive oxygen species (mROS) play a crucial physiological role in intracellular signalling. However, high levels of ROS can overwhelm antioxidant defences and lead to detrimental modifications in protein, lipid and DNA structure and function. Ischaemia-reperfusion injury is a multifaceted pathological state characterised by excessive production of mROS. There is a significant clinical need for therapies mitigating mitochondrial oxidative stress. To date, a variety of strategies have been investigated, ranging from enhancing antioxidant reserve capacity to metabolism reduction. While success has been achieved in non-clinical models, no intervention has yet successfully transitioned into routine clinical practice. In this article, we explore the different strategies investigated and discuss the possible reasons for the lack of translation.
Collapse
Affiliation(s)
- Khalid Alotaibi
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, U.K
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Nishkantha Arulkumaran
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, U.K
| | - Alex Dyson
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, U.K
- Centre for Pharmaceutical Medicine Research, Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, U.K
| |
Collapse
|
2
|
Eberle MJ, Thorkelsson AB, Liddle LJ, Almekhlafi M, Colbourne F. Longer Periods of Hypothermia Provide Greater Protection Against Focal Ischemia: A Systematic Review of Animal Studies Manipulating Treatment Duration. Ther Hypothermia Temp Manag 2024; 14:144-151. [PMID: 37788401 DOI: 10.1089/ther.2023.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
Decades of animal research show therapeutic hypothermia (TH) to be potently neuroprotective after cerebral ischemic injuries. While there have been some translational successes, clinical efficacy after ischemic stroke is unclear. One potential reason for translational failures could be insufficient optimization of dosing parameters. In this study, we conducted a systematic review of the PubMed database to identify all preclinical controlled studies that compared multiple TH durations following focal ischemia, with treatment beginning at least 1 hour after ischemic onset. Six studies met our inclusion criteria. In these six studies, six of seven experiments demonstrated an increase in cerebroprotection at the longest duration tested. The average effect size (mean Cohen's d ± 95% confidence interval) at the shortest and longest durations was 0.4 ± 0.3 and 1.9 ± 1.1, respectively. At the longest durations, this corresponded to percent infarct volume reductions between 31.2% and 83.9%. Our analysis counters previous meta-analytic findings that there is no relationship, or an inverse relationship between TH duration and effect size. However, underreporting often led to high or unclear risks of bias for each study as gauged by the SYRCLE Risk of Bias tool. We also found a lack of investigations of the interactions between duration and other treatment considerations (e.g., method, delay, and ischemic severity). With consideration of methodological limitations, an understanding of the relationships between treatment parameters is necessary to determine proper "dosage" of TH, and should be further studied, considering clinical failures that contrast with strong cerebroprotective results in most animal studies.
Collapse
Affiliation(s)
- Megan J Eberle
- Neuroscience and Mental Health Institute, and University of Alberta, Edmonton, Canada
| | | | - Lane J Liddle
- Department of Psychology, University of Alberta, Edmonton, Canada
| | - Mohammed Almekhlafi
- Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Calgary, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, and University of Alberta, Edmonton, Canada
- Department of Psychology, University of Alberta, Edmonton, Canada
| |
Collapse
|
3
|
Díaz-Peregrino R, Kentar M, Trenado C, Sánchez-Porras R, Albiña-Palmarola P, Ramírez-Cuapio FL, San-Juan D, Unterberg A, Woitzik J, Santos E. The neurophysiological effect of mild hypothermia in gyrencephalic brains submitted to ischemic stroke and spreading depolarizations. Front Neurosci 2024; 18:1302767. [PMID: 38567280 PMCID: PMC10986791 DOI: 10.3389/fnins.2024.1302767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Objective Characterize the neurophysiological effects of mild hypothermia on stroke and spreading depolarizations (SDs) in gyrencephalic brains. Methods Left middle cerebral arteries (MCAs) of six hypothermic and six normothermic pigs were permanently occluded (MCAo). Hypothermia began 1 h after MCAo and continued throughout the experiment. ECoG signals from both frontoparietal cortices were recorded. Five-minute ECoG epochs were collected 5 min before, at 5 min, 4, 8, 12, and 16 h after MCAo, and before, during, and after SDs. Power spectra were decomposed into fast (alpha, beta, and gamma) and slow (delta and theta) frequency bands. Results In the vascular insulted hemisphere under normothermia, electrodes near the ischemic core exhibited power decay across all frequency bands at 5 min and the 4th hour after MCAo. The same pattern was registered in the two furthest electrodes at the 12th and 16th hour. When mild hypothermia was applied in the vascular insulted hemispheres, the power decay was generalized and seen even in electrodes with uncompromised blood flow. During SD analysis, hypothermia maintained increased delta and beta power during the three phases of SDs in the furthest electrode from the ischemic core, followed by the second furthest and third electrode in the beta band during preSD and postSD segments. However, in hypothermic conditions, the third electrode showed lower delta, theta, and alpha power. Conclusion Mild hypothermia attenuates all frequency bands in the vascularly compromised hemisphere, irrespective of the cortical location. During SD formation, it preserves power spectra more significantly in electrodes further from the ischemic core.
Collapse
Affiliation(s)
- Roberto Díaz-Peregrino
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Modar Kentar
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
- Departement of Neurosurgery, Städtisches Klinikum Braunschweig gGmbH, Braunschweig, Germany
| | - Carlos Trenado
- Heinrich Heine University, Medical Faculty, Institute of Clinical Neuroscience and Medical Psychology, Düsseldorf, Germany
- Institute for the Future of Education Europe, Tecnológico de Monterrey, Cantabria, Spain
| | - Renán Sánchez-Porras
- Department of Neurosurgery, Evangelisches Krankenhaus, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Pablo Albiña-Palmarola
- Neuroradiologische Klinik, Klinikum Stuttgart, Stuttgart, Germany
- Medizinische Fakultät, Universität Duisburg-Essen, Essen, Germany
- Department of Anatomy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco L. Ramírez-Cuapio
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Daniel San-Juan
- Epilepsy Clinic, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Edgar Santos
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
- Department of Neurosurgery, Evangelisches Krankenhaus, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
4
|
Li M, Gao Y, Jiang M, Zhang H, Zhang Y, Wu Y, Zhou W, Wu D, Wu C, Wu L, Bao L, Ge X, Qi Z, Wei M, Li A, Ding Y, Zhang J, Pan G, Wu Y, Cheng Y, Zheng Y, Ji X. Dual-sized hollow particle incorporated fibroin thermal insulating coatings on catheter for cerebral therapeutic hypothermia. Bioact Mater 2023; 26:116-127. [PMID: 36879558 PMCID: PMC9984786 DOI: 10.1016/j.bioactmat.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 02/19/2023] [Accepted: 02/19/2023] [Indexed: 02/27/2023] Open
Abstract
Selective endovascular hypothermia has been used to provide cooling-induced cerebral neuroprotection, but current catheters do not support thermally-insulated transfer of cold infusate, which results in an increased exit temperature, causes hemodilution, and limits its cooling efficiency. Herein, air-sprayed fibroin/silica-based coatings combined with chemical vapor deposited parylene-C capping film was prepared on catheter. This coating features in dual-sized-hollow-microparticle incorporated structures with low thermal conductivity. The infusate exit temperature is tunable by adjusting the coating thickness and infusion rate. No peeling or cracking was observed on the coatings under bending and rotational scenarios in the vascular models. Its efficiency was verified in a swine model, and the outlet temperature of coated catheter (75 μm thickness) was 1.8-2.0 °C lower than that of the uncoated one. This pioneering work on catheter thermal insulation coatings may facilitate the clinical translation of selective endovascular hypothermia for neuroprotection in patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Ming Li
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuan Gao
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, 100191, China
| | - Miaowen Jiang
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Hongkang Zhang
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yang Zhang
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yan Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wenhao Zhou
- Shaanxi Key Laboratory of Biomedical Metallic Materials, Northwest Institute for Nonferrous Metal Research, Xi'an, 710016, China
| | - Di Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Chuanjie Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Longfei Wu
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Luzi Bao
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiaoxiao Ge
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Zhengfei Qi
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ming Wei
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ang Li
- Department of Biomedical Engineering, Columbia University, New York City, NY, 10027, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Jicheng Zhang
- Gong Yi Van-research Innovation Composite Material Co. Ltd, Zheng Zhou, 451299, China
| | - Guangzhen Pan
- Gong Yi Van-research Innovation Composite Material Co. Ltd, Zheng Zhou, 451299, China
| | - Yu Wu
- Gong Yi Van-research Innovation Composite Material Co. Ltd, Zheng Zhou, 451299, China
| | - Yan Cheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xunming Ji
- China-America Institute of Neuroscience and Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.,School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, 100191, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| |
Collapse
|
5
|
Kentar M, Ramirez-Cuapio FL, Gutiérrez-Herrera MA, Sanchez-Porras R, Díaz-Peregrino R, Holzwarth N, Maier-Hein L, Woitzik J, Santos E. Mild hypothermia reduces spreading depolarizations and infarct size in a swine model. J Cereb Blood Flow Metab 2023; 43:999-1009. [PMID: 36722153 PMCID: PMC10196741 DOI: 10.1177/0271678x231154604] [Citation(s) in RCA: 3] [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: 08/18/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 02/02/2023]
Abstract
Spreading depolarizations (SDs) have been linked to infarct volume expansion following ischemic stroke. Therapeutic hypothermia provides a neuroprotective effect after ischemic stroke. This study aimed to evaluate the effect of hypothermia on the propagation of SDs and infarct volume in an ischemic swine model. Through left orbital exenteration, middle cerebral arteries were surgically occluded (MCAo) in 16 swine. Extensive craniotomy and durotomy were performed. Six hypothermic and five normothermic animals were included in the analysis. An intracranial temperature probe was placed right frontal subdural. One hour after ischemic onset, mild hypothermia was induced and eighteen hours of electrocorticographic (ECoG) and intrinsic optical signal (IOS) recordings were acquired. Postmortem, 4 mm-thick slices were stained with 2,3,5-triphenyltetrazolium chloride to estimate the infarct volume. Compared to normothermia (36.4 ± 0.4°C), hypothermia (32.3 ± 0.2°C) significantly reduced the frequency and expansion of SDs (ECoG: 3.5 ± 2.1, 73.2 ± 5.2% vs. 1.0 ± 0.7, 41.9 ± 21.8%; IOS 3.9 ± 0.4, 87.6 ± 12.0% vs. 1.4 ± 0.7, 67.7 ± 8.3%, respectively). Further, infarct volume among hypothermic animals (23.2 ± 1.8% vs. 32.4 ± 2.5%) was significantly reduced. Therapeutic hypothermia reduces infarct volume and the frequency and expansion of SDs following cerebral ischemia.
Collapse
Affiliation(s)
- Modar Kentar
- Department of Neurosurgery,
University of Heidelberg, Heidelberg, Germany
| | | | | | - Renan Sanchez-Porras
- Department of Neurosurgery,
Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg,
Oldenburg, Germany
| | | | - Niklas Holzwarth
- Division of Intelligent Medical
Systems, German Cancer Research Center, Heidelberg, Germany
| | - Lena Maier-Hein
- Division of Intelligent Medical
Systems, German Cancer Research Center, Heidelberg, Germany
| | - Johannes Woitzik
- Department of Neurosurgery,
Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg,
Oldenburg, Germany
| | - Edgar Santos
- Department of Neurosurgery,
University of Heidelberg, Heidelberg, Germany
- Department of Neurosurgery,
Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg,
Oldenburg, Germany
| |
Collapse
|
6
|
Battaglini D, da Silva AL, Felix NS, Rodrigues G, Antunes MA, Rocha NN, Capelozzi VL, Morales MM, Cruz FF, Robba C, Silva PL, Pelosi P, Rocco PRM. Mild hypothermia combined with dexmedetomidine reduced brain, lung, and kidney damage in experimental acute focal ischemic stroke. Intensive Care Med Exp 2022; 10:53. [PMID: 36529842 PMCID: PMC9760586 DOI: 10.1186/s40635-022-00481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sedatives and mild hypothermia alone may yield neuroprotective effects in acute ischemic stroke (AIS). However, the impact of this combination is still under investigation. We compared the effects of the combination of mild hypothermia or normothermia with propofol or dexmedetomidine on brain, lung, and kidney in experimental AIS. AIS-induced Wistar rats (n = 30) were randomly assigned, after 24 h, to normothermia or mild hypothermia (32-35 °C) with propofol or dexmedetomidine. Histologic injury score and molecular biomarkers were evaluated not only in brain, but also in lung and kidney. Hemodynamics, ventilatory parameters, and carotid Doppler ultrasonography were analyzed for 60 min. RESULTS In brain: (1) hypothermia compared to normothermia, regardless of sedative, decreased tumor necrosis factor (TNF)-α expression and histologic injury score; (2) normothermia + dexmedetomidine reduced TNF-α and histologic injury score compared to normothermia + propofol; (3) hypothermia + dexmedetomidine increased zonula occludens-1 expression compared to normothermia + dexmedetomidine. In lungs: (1) hypothermia + propofol compared to normothermia + propofol reduced TNF-α and histologic injury score; (2) hypothermia + dexmedetomidine compared to normothermia + dexmedetomidine reduced histologic injury score. In kidneys: (1) hypothermia + dexmedetomidine compared to normothermia + dexmedetomidine decreased syndecan expression and histologic injury score; (2) hypothermia + dexmedetomidine compared to hypothermia + propofol decreased histologic injury score. CONCLUSIONS In experimental AIS, the combination of mild hypothermia with dexmedetomidine reduced brain, lung, and kidney damage.
Collapse
Affiliation(s)
- Denise Battaglini
- grid.410345.70000 0004 1756 7871Anesthesiology and Critical Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy ,grid.5841.80000 0004 1937 0247Department of Medicine, University of Barcelona, 08007 Barcelona, Spain ,grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Adriana Lopes da Silva
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Nathane Santanna Felix
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Gisele Rodrigues
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Mariana Alves Antunes
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Nazareth Novaes Rocha
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil ,grid.411173.10000 0001 2184 6919Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, 24220-900 Brazil
| | - Vera Luiza Capelozzi
- grid.11899.380000 0004 1937 0722Department of Pathology, University of São Paolo, São Paolo, 05508-060 Brazil
| | - Marcelo Marcos Morales
- grid.8536.80000 0001 2294 473XLaboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901 Brazil
| | - Fernanda Ferreira Cruz
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Chiara Robba
- grid.410345.70000 0004 1756 7871Anesthesiology and Critical Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy ,grid.5606.50000 0001 2151 3065Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Pedro Leme Silva
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil
| | - Paolo Pelosi
- grid.410345.70000 0004 1756 7871Anesthesiology and Critical Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, 16132 Genoa, Italy ,grid.5606.50000 0001 2151 3065Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia Rieken Macedo Rocco
- grid.8536.80000 0001 2294 473XLaboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ 21941-902 Brazil ,grid.452991.20000 0000 8484 4876Rio de Janeiro Network On Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Mattingly TK, McDavid A, Wolf A, Lieber G, Solar R, Lee D, Lownie SP. The Complex Relationship Between Cooling Parameters and Neuroprotection in a Model of Selective Hypothermia. Front Neurol 2022; 13:874701. [PMID: 35547387 PMCID: PMC9081928 DOI: 10.3389/fneur.2022.874701] [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: 02/12/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background Hypothermia remains the best studied neuroprotectant. Despite extensive positive large and small animal data, side effects continue to limit human applications. Selective hypothermia is an efficient way of applying neuroprotection to the brain without the systemic complications of global hypothermia. However, optimal depth and duration of therapeutic hypothermia are still unknown. We analyzed a large animal cohort study of selective hypothermia for statistical relationships between depth or duration of hypothermia and the final stroke volume. Methods A cohort of 30 swine stroke subjects provided the dataset for normothermic and selective hypothermic animals. Hypothermic parameters including duration, temperature nadir, and an Area Under the Curve measurement for 34 and 30°C were correlated with the final infarct volumes measured by MRI and histology. Results Between group comparisons continue to demonstrate a reduction in infarct volume with selective hypothermia. Histologically-derived infarct volumes were 1.2 mm3 smaller in hypothermia-treated pigs (P = 0.04) and showed a similar, but non-significant reduction in MRI (P = 0.15). However, within the selective hypothermia group, more intense cooling, as measured through increased AUC 34 and decreased temperature nadir was associated with larger infarct proportions by MRI [Pearson's r = 0.48 (p = 0.05) and r = -0.59 (p = 0.01), respectively]. Reevaluation of the entire cohort with quadratic regression demonstrated a U-shaped pattern, wherein the average infarct proportion was minimized at 515 degree-minutes (AUC34) of cooling, and increased thereafter. In a single case of direct brain tissue oxygen monitoring during selective hypothermia, brain tissue oxygen strongly correlated with brain temperature reduction over the course of selective hypothermia to 23°C. Conclusions In a large animal model of selective hypothermia applied to focal ischemia, there is a non-monotone relationship between duration and depth of hypothermia and stroke volume reduction. This suggests a limit to depth or duration of selective hypothermia for optimal neuroprotection. Further research is required to delineate more precise depth and duration limits for selective hypothermia.
Collapse
Affiliation(s)
- Thomas K Mattingly
- Department of Neurosurgery, University of Rochester, Rochester, NY, United States
| | - Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Amparo Wolf
- Department of Neurosurgery, Health Sciences North, Sudbury, ON, Canada
| | - Glen Lieber
- ThermopeutiX, Inc., San Diego, CA, United States
| | - Ronald Solar
- ThermopeutiX, Inc., San Diego, CA, United States
| | - Donald Lee
- Department of Medical Imaging, London Health Science Centre, London, ON, Canada
| | - Stephen P Lownie
- Division of Neurosurgery, Halifax Infirmary, Halifax, NS, Canada
| |
Collapse
|
8
|
Omileke D, Pepperall D, Bothwell SW, Mackovski N, Azarpeykan S, Beard DJ, Coupland K, Patabendige A, Spratt NJ. Ultra-Short Duration Hypothermia Prevents Intracranial Pressure Elevation Following Ischaemic Stroke in Rats. Front Neurol 2021; 12:684353. [PMID: 34616350 PMCID: PMC8488292 DOI: 10.3389/fneur.2021.684353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
There is a transient increase in intracranial pressure (ICP) 18–24 h after ischaemic stroke in rats, which is prevented by short-duration hypothermia using rapid cooling methods. Clinical trials of long-duration hypothermia have been limited by feasibility and associated complications, which may be avoided by short-duration cooling. Animal studies have cooled faster than is achievable in patients. We aimed to determine whether gradual cooling at a rate of 2°C/h to 33°C or 1°C/h to 34.5°C, with a 30 min duration at target temperatures, prevented ICP elevation and reduced infarct volume in rats. Transient middle cerebral artery occlusion was performed, followed by gradual cooling to target temperature. Hypothermia to 33°C prevented significant ICP elevation (hypothermia ΔICP = 1.56 ± 2.26 mmHg vs normothermia ΔICP = 8.93 ± 4.82 mmHg; p = 0.02) and reduced infarct volume (hypothermia = 46.4 ± 12.3 mm3 vs normothermia = 85.0 ± 17.5 mm3; p = 0.01). Hypothermia to 34.5°C did not significantly prevent ICP elevation or reduce infarct volume. We showed that gradual cooling to 33°C, at cooling rates achievable in patients, had the same ICP preventative effect as traditional rapid cooling methods. This suggests that this paradigm could be translated to prevent delayed ICP rise in stroke patients.
Collapse
Affiliation(s)
- Daniel Omileke
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Debbie Pepperall
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Steven W Bothwell
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Nikolce Mackovski
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Sara Azarpeykan
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Daniel J Beard
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Kirsten Coupland
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Adjanie Patabendige
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Neil J Spratt
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia.,Department of Neurology, John Hunter Hospital, Hunter New England Local Health District, New Lambton, NSW, Australia
| |
Collapse
|
9
|
Liddle LJ, Kalisvaart ACJ, Abrahart AH, Almekhlafi M, Demchuk A, Colbourne F. Targeting focal ischemic and hemorrhagic stroke neuroprotection: Current prospects for local hypothermia. J Neurochem 2021; 160:128-144. [PMID: 34496050 DOI: 10.1111/jnc.15508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/01/2021] [Accepted: 09/05/2021] [Indexed: 01/17/2023]
Abstract
Therapeutic hypothermia (TH) has applications dating back millennia. In modern history, however, TH saw its importation into medical practice where investigations have demonstrated that TH is efficacious in ischemic insults, notably cardiac arrest and hypoxic-ischemic encephalopathy. As well, studies have been undertaken to investigate whether TH can provide benefit in focal stroke (i.e., focal ischemia and intracerebral hemorrhage). However, clinical studies have encountered various challenges with induction and maintenance of post-stroke TH. Most clinical studies have attempted to use body-wide cooling protocols, commonly hindered by side effects that can worsen post-stroke outcomes. Some of the complications and difficulties with systemic TH can be circumvented by using local hypothermia (LH) methods. Additional advantages include the potential for lower target temperatures to be achieved and faster TH induction rates with LH. This systematic review summarizes the body of clinical and preclinical LH focal stroke studies and raises key points to consider for future LH research. We conclude with an overview of LH neuroprotective mechanisms and a comparison of LH mechanisms with those observed with systemic TH. Overall, whereas many LH studies have been conducted preclinically in the context of focal ischemia, insufficient work has been done in intracerebral hemorrhage. Furthermore, key translational studies have yet to be done in either stroke subtype (e.g., varied models and time-to-treat, studies considering aged animals or animals with co-morbidities). Few clinical LH investigations have been performed and the optimal LH parameters to achieve neuroprotection are unknown.
Collapse
Affiliation(s)
- Lane J Liddle
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Ashley H Abrahart
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
10
|
Ageing as a risk factor for cerebral ischemia: Underlying mechanisms and therapy in animal models and in the clinic. Mech Ageing Dev 2020; 190:111312. [PMID: 32663480 DOI: 10.1016/j.mad.2020.111312] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Age is the only one non-modifiable risk of cerebral ischemia. Advances in stroke medicine and behavioral adaptation to stroke risk factors and comorbidities was successful in decreasing stroke incidence and increasing the number of stroke survivors in western societies. Comorbidities aggravates the outcome after cerebral ischemia. However, due to the increased in number of elderly, the incidence of stroke has increased again paralleled by an increase in the number of stroke survivors, many with severe disabilities, that has led to an increased economic and social burden in society. Animal models of stroke often ignore age and comorbidities frequently associated with senescence. This might explain why drugs working nicely in animal models fail to show efficacy in stroke survivors. Since stroke afflicts mostly the elderly comorbid patients, it is highly desirable to test the efficacy of stroke therapies in an appropriate animal stroke model. Therefore, in this review, we make parallels between animal models of stroke und clinical data and summarize the impact of ageing and age-related comorbidities on stroke outcome.
Collapse
|
11
|
Koehn J, Wang R, de Rojas Leal C, Kallmünzer B, Winder K, Köhrmann M, Kollmar R, Schwab S, Hilz MJ. Neck cooling induces blood pressure increase and peripheral vasoconstriction in healthy persons. Neurol Sci 2020; 41:2521-2529. [PMID: 32219592 PMCID: PMC8197712 DOI: 10.1007/s10072-020-04349-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/16/2020] [Indexed: 12/18/2022]
Abstract
Introduction Noninvasive temperature modulation by localized neck cooling might be desirable in the prehospital phase of acute hypoxic brain injuries. While combined head and neck cooling induces significant discomfort, peripheral vasoconstriction, and blood pressure increase, localized neck cooling more selectively targets blood vessels that supply the brain, spares thermal receptors of the face and skull, and might therefore cause less discomfort cardiovascular side effects compared to head- and neck cooling. The purpose of this study is to assess the effects of noninvasive selective neck cooling on cardiovascular parameters and cerebral blood flow velocity (CBFV). Methods Eleven healthy persons (6 women, mean age 42 ± 11 years) underwent 90 min of localized dorsal and frontal neck cooling (EMCOOLS Brain.Pad™) without sedation. Before and after cooling onset, and after every 10 min of cooling, we determined rectal, tympanic, and neck skin temperatures. Before and after cooling onset, after 60- and 90-min cooling, we monitored RR intervals (RRI), systolic, diastolic blood pressures (BPsys, BPdia), laser Doppler skin blood flow (SBF) at the index finger pulp, and CBFV at the proximal middle cerebral artery (MCA). We compared values before and during cooling by analysis of variance for repeated measurements with post hoc analysis (significance: p < 0.05). Results Neck skin temperature dropped significantly by 9.2 ± 4.5 °C (minimum after 40 min), while tympanic temperature decreased by only 0.8 ± 0.4 °C (minimum after 50 min), and rectal temperature by only 0.2 ± 0.3 °C (minimum after 60 min of cooling). Index finger SBF decreased (by 83.4 ± 126.0 PU), BPsys and BPdia increased (by 11.2 ± 13.1 mmHg and 8.0 ± 10.1 mmHg), and heart rate slowed significantly while MCA-CBFV remained unchanged during cooling. Conclusions While localized neck cooling prominently lowered neck skin temperature, it had little effect on tympanic temperature but significantly increased BP which may have detrimental effects in patients with acute brain injuries.
Collapse
Affiliation(s)
- Julia Koehn
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Ruihao Wang
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Carmen de Rojas Leal
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Bernd Kallmünzer
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Klemens Winder
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Martin Köhrmann
- Department of Neurology, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Rainer Kollmar
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany.,Department of Neurology, General Hospital Darmstadt, Grafenstr. 9, 64283, Darmstadt, Germany
| | - Stefan Schwab
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Max J Hilz
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany. .,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Wu L, Wu D, Yang T, Xu J, Chen J, Wang L, Xu S, Zhao W, Wu C, Ji X. Hypothermic neuroprotection against acute ischemic stroke: The 2019 update. J Cereb Blood Flow Metab 2020; 40:461-481. [PMID: 31856639 PMCID: PMC7026854 DOI: 10.1177/0271678x19894869] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 02/06/2023]
Abstract
Acute ischemic stroke is a leading cause of death and disability worldwide. Therapeutic hypothermia has long been considered as one of the most robust neuroprotective strategies. Although the neuroprotective effects of hypothermia have only been confirmed in patients with global cerebral ischemia after cardiac arrest and in neonatal hypoxic ischemic encephalopathy, establishing standardized protocols and strictly controlling the key parameters may extend its application in other brain injuries, such as acute ischemic stroke. In this review, we discuss the potential neuroprotective effects of hypothermia, its drawbacks evidenced in previous studies, and its potential clinical application for acute ischemic stroke especially in the era of reperfusion. Based on the different conditions between bench and bedside settings, we demonstrate the importance of vascular recanalization for neuroprotection of hypothermia by analyzing numerous literatures regarding hypothermia in focal cerebral ischemia. Then, we make a thorough analysis of key parameters of hypothermia and introduce novel hypothermic therapies. We advocate in favor of the process of clinical translation of intra-arterial selective cooling infusion in the era of reperfusion and provide insights into the prospects of hypothermia in acute ischemic stroke.
Collapse
Affiliation(s)
- Longfei Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jin Xu
- Department of Library, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jian Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Luling Wang
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chuanjie Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Neugebauer H, Schneider H, Bösel J, Hobohm C, Poli S, Kollmar R, Sobesky J, Wolf S, Bauer M, Tittel S, Beyersmann J, Woitzik J, Heuschmann PU, Jüttler E. Outcomes of Hypothermia in Addition to Decompressive Hemicraniectomy in Treatment of Malignant Middle Cerebral Artery Stroke: A Randomized Clinical Trial. JAMA Neurol 2020; 76:571-579. [PMID: 30657812 DOI: 10.1001/jamaneurol.2018.4822] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Importance Moderate hypothermia in addition to early decompressive hemicraniectomy has been suggested to further reduce mortality and improve functional outcome in patients with malignant middle cerebral artery (MCA) stroke. Objective To investigate whether moderate hypothermia vs standard treatment after early hemicraniectomy reduces mortality at day 14 in patients with malignant MCA stroke. Design, Setting, and Participants This randomized clinical trial recruited patients from August 2011 through September 2015 at 6 German university hospitals with dedicated neurointensive care units. Of the patients treated with hemicraniectomy and assessed for eligibility, patients were randomly assigned to either standard care or moderate hypothermia. Data analysis was completed from December 2016 to June 2018. Interventions Moderate hypothermia (temperature, 33.0 ± 1.0°C) was maintained for at least 72 hours immediately after hemicraniectomy. Main Outcomes and Measures The primary outcome was mortality rate at day 14 compared with the Fisher exact test and expressed as odds ratio (ORs) with 95% CIs. Rates of patients with serious adverse events were estimated for the period of the first 14 days after hemicraniectomy and 12 months of follow-up. Secondary outcome measures included functional outcome at 12 months. Results Of the 50 study participants, 24 were assigned to standard care and 26 to moderate hypothermia. Twenty-eight were male (56%); the mean (SD) patient age was 51.3 (6.6) years. Recruitment was suspended for safety concerns: 12 of 26 patients (46%) in the hypothermia group and 7 of 24 patients (29%) receiving standard care had at least 1 serious adverse event within 14 days (OR, 2.05 [95% CI, 0.56-8.00]; P = .26); after 12 months, rates of serious adverse events were 80% (n = 20 of 25) in the hypothermia group and 43% (n = 10 of 23) in the standard care group (hazard ratio, 2.54 [95% CI, 1.29-5.00]; P = .005). The mortality rate at day 14 was 19% (5 of 26 patients) in the hypothermia group and 13% (3 of 24 patients) in the group receiving standard care (OR, 1.65 [95% CI, 0.28-12.01]; P = .70). There was no significant difference regarding functional outcome after 12 months of follow-up. Interpretation In patients with malignant MCA stroke, moderate hypothermia early after hemicraniectomy did not improve mortality and functional outcome compared with standard care, but may cause serious harm in this specific setting. Trial Registration http://www.drks.de, identifier DRKS00000623.
Collapse
Affiliation(s)
- Hermann Neugebauer
- Department of Neurology, RKU-University and Rehabilitation Hospitals Ulm, University of Ulm, Ulm, Germany
| | - Hauke Schneider
- Department of Neurology, University Hospital, Technische Universität Dresden, Dresden, Germany.,Department of Neurology, Universitätsklinikum Augsburg, Augsburg, Germany
| | - Julian Bösel
- Department of Neurology, University of Heidelberg, Heidelberg, Germany.,Department of Neurology, Klinikum Kassel, Kassel, Germany
| | - Carsten Hobohm
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Klinikum Saalekreis, Merseburg, Germany
| | - Sven Poli
- Department of Neurology, University of Tübingen, Tübingen, Germany
| | - Rainer Kollmar
- Department of Neurology, University of Erlangen, Erlangen, Germany.,Department of Neurology and Neurointensive Care, Klinikum Darmstadt, Darmstadt, Germany
| | - Jan Sobesky
- Department of Neurology and Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Johanna-Etienne-Krankenhaus Neuss, Neuss, Germany
| | - Stefan Wolf
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Miriam Bauer
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sascha Tittel
- Institute of Statistics, University of Ulm, Ulm, Germany
| | - Jan Beyersmann
- Institute of Statistics, University of Ulm, Ulm, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter U Heuschmann
- Institute for Clinical Epidemiology and Biometry, Comprehensive Heart Failure Centre, University of Würzburg, Würzburg, Germany.,Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - Eric Jüttler
- Department of Neurology, RKU-University and Rehabilitation Hospitals Ulm, University of Ulm, Ulm, Germany.,Department of Neurology, Ostalb-Klinikum Aalen, Aalen, Germany
| |
Collapse
|
14
|
Katica-Mulalic A, Suljic E, Begic E, Mukanovic-Alihodzic A, Straus S, Feto A, Dedovic Z, Gojak R. Effect of Therapeutic Hypothermia on Liver Enzymes in Patients With Stroke. Med Arch 2020; 74:470-473. [PMID: 33603273 PMCID: PMC7879342 DOI: 10.5455/medarh.2020.74.470-473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction: A promising strategy that can lead to longer brain cell survival after an acute stroke is therapeutic hypothermia. It represents a controlled decrease in body temperature for therapeutic reasons. It is increasingly represented as a therapeutic option and is one of the most challenging treatments that improves neurological recovery and treatment outcome in patients with acute stroke. Aim: To examine the effect of therapeutic hypothermia on liver enzymes in patients with diagnosis of stroke. Methods: A total of 101 patients diagnosed with acute stroke were treated. The first group (n=40) were treated with conventional treatment and therapeutic hypothermia, while the second group (n=61) only with conventional treatment. Cooling of the body to a target body temperature of 34°C to 35°C was performed for up to 24 hours. Outcome (survival or death) of treatment was monitored, degree of disability was determined by National Institutes of Health Stroke Scale (NIHSS) and assessment of consciousness using the Glasgow Coma Scale (GCS). Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) values were taken at admission, after 24 hours, and were monitored upon discharge. Results: There was a significant difference in AST values at admission relative to disease outcome (p = 0.002), as well as for ALT (p = 0.008). In patients treated with therapeutic hypothermia, mean AST values decreased after 24 hours (32.50 to 31.00 IU/mL) as well as ALT values (27.50 to 26.50 IU/mL), without statistical significance. In the group of subjects who survived with sequela, AST values correlated with GCS (rho = -0.489; p = 0.002) and NIHSS (rho = 0.492; p = 0.003), ALT values correlated with GCS (rho = -0.356; p = 0.03) but not with NIHSS. Conclusion: AST and ALT values at admission correlate with the severity of the clinical picture. Therapeutic hypothermia is hepatoprotective and lowers AST and ALT values.
Collapse
Affiliation(s)
- Amela Katica-Mulalic
- Clinic Anesthesiology and Resuscitation, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Enra Suljic
- Department for Science, Teaching and Clinical Trials, Clinical Centre University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Edin Begic
- Department of Cardiology, General Hospital «Prim. dr. Abdulah Nakas», Sarajevo, Bosnia and Herzegovina
| | - Azra Mukanovic-Alihodzic
- Clinic Anesthesiology and Resuscitation, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Slavenka Straus
- Clinic for Cardiovascular Surgery, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Amila Feto
- Clinic Anesthesiology and Resuscitation, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Zenaida Dedovic
- Clinic Anesthesiology and Resuscitation, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Refet Gojak
- Clinic for Infectious Diseases, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
15
|
Kalisvaart ACJ, Prokop BJ, Colbourne F. Hypothermia: Impact on plasticity following brain injury. Brain Circ 2019; 5:169-178. [PMID: 31950092 PMCID: PMC6950515 DOI: 10.4103/bc.bc_21_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
Therapeutic hypothermia (TH) is a potent neuroprotectant against multiple forms of brain injury, but in some cases, prolonged cooling is needed. Such cooling protocols raise the risk that TH will directly or indirectly impact neuroplasticity, such as after global and focal cerebral ischemia or traumatic brain injury. TH, depending on the depth and duration, has the potential to broadly affect brain plasticity, especially given the spatial, temporal, and mechanistic overlap with the injury processes that cooling is used to treat. Here, we review the current experimental and clinical evidence to evaluate whether application of TH has any adverse or positive effects on postinjury plasticity. The limited available data suggest that mild TH does not appear to have any deleterious effect on neuroplasticity; however, we emphasize the need for additional high-quality preclinical and clinical work in this area.
Collapse
|
16
|
Wang D, Huang Z, Li L, Yuan Y, Xiang L, Wu X, Ni C, Yu W. Intracarotid cold saline infusion contributes to neuroprotection in MCAO‑induced ischemic stroke in rats via serum and glucocorticoid‑regulated kinase 1. Mol Med Rep 2019; 20:3942-3950. [PMID: 31485662 DOI: 10.3892/mmr.2019.10599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 07/16/2019] [Indexed: 11/05/2022] Open
Abstract
Intracarotid cold saline infusion (ICSI) brings about neuroprotective effects in ischemic stroke. However, the involvement of serum and glucocorticoid‑regulated kinase 1 (SGK1) in the underlying mechanism of ICSI is not fully understood; therefore, we used the rat middle cerebral artery occlusion (MCAO) model to investigate the neuroprotective effects of ICSI on ischemic stroke in rats, as well as the involvement of SGK1 in these effects. ICSI decreased infarct size and brain swelling, as determined by 2,3,5‑triphenyltetrazolium chloride staining and the dry‑wet weight method, respectively. The results of terminal deoxynucleotidyl transferase mediated nick end labeling (TUNEL) and Nissl staining showed that ICSI also suppressed apoptosis and increased the relative integral optical density (IOD) values of Nissl bodies in the rat MCAO model. Regarding the mechanism, the results of immunohistochemistry and western blotting revealed that ICSI upregulated SGK1 expression and downregulated beclin‑1 and LC‑3 expression in the rat MCAO model. In addition, SGK1 knockdown increased ICSI‑mediated infarct size and brain swelling, promoted apoptosis, and reduced the IOD values of Nissl bodies in the rat MCAO model. In addition, we found that SGK1 knockdown upregulated beclin‑1 and LC‑3 expression mediated by ICSI. Overall, ICSI had a neuroprotective effect on ischemic stroke after reperfusion by upregulating SGK1 and inhibiting autophagy.
Collapse
Affiliation(s)
- Dazhi Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Zhi Huang
- Department of Interventional Radiology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, P.R. China
| | - Lei Li
- Department of General Courses, People's Armed College of Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Yingnan Yuan
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Lei Xiang
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Xiaowen Wu
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wenfeng Yu
- Key Laboratory of Molecular Biology, Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
17
|
Hypothermia in the Neurocritical Care Unit: Physiology and Applications. Neurocrit Care 2019. [DOI: 10.1017/9781107587908.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
18
|
|
19
|
Tauchi M, Tejada de Rink MM, Fujioka H, Okayama S, Nakamura KI, Dietel B, Achenbach S, Kollmar R, Schwab S, Ushijima K, Harada H. Targeted Temperature Management: Peltier's Element-Based Focal Brain Cooling Protects Penumbra Neurons from Progressive Damage in Experimental Cerebral Ischemia. Ther Hypothermia Temp Manag 2018; 8:225-233. [DOI: 10.1089/ther.2017.0055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Miyuki Tauchi
- Department of Neurology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Molecular Neurology, and Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Medicine 2–Cardiology and Angiology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maria Mercedes Tejada de Rink
- Department of Anesthesiology, Kurume University School of Medicine, Kurume, Japan
- Neuroanesthesia Research Laboratory, Cognitive and Molecular Institute of Brain Diseases, Kurume University School of Medicine, Kurume, Japan
| | - Hiroshi Fujioka
- Department of Neurosurgery, Kanmon Medical Center, National Hospital Organization (NHO), Yamaguchi, Japan
| | - Satoko Okayama
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Kei-ichiro Nakamura
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Barbara Dietel
- Department of Medicine 2–Cardiology and Angiology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stephan Achenbach
- Department of Medicine 2–Cardiology and Angiology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Kollmar
- Department of Neurology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefan Schwab
- Department of Neurology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kazuo Ushijima
- Department of Anesthesiology, Kurume University School of Medicine, Kurume, Japan
| | - Hideki Harada
- Department of Anesthesiology, Kurume University School of Medicine, Kurume, Japan
- Neuroanesthesia Research Laboratory, Cognitive and Molecular Institute of Brain Diseases, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
20
|
Bobinger T, Burkardt P, B Huttner H, Manaenko A. Programmed Cell Death after Intracerebral Hemorrhage. Curr Neuropharmacol 2018; 16:1267-1281. [PMID: 28571544 PMCID: PMC6251052 DOI: 10.2174/1570159x15666170602112851] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 06/01/2017] [Indexed: 01/01/2023] Open
Abstract
Background: Intracerebral hemorrhage (ICH) accounts for up to 15% of all strokes and is characterized by high rates of mortality and morbidity. The post-ICH brain injury can be distinguished in 1) primary, which are caused by disrup-tion and mechanical deformation of brain tissue due to hematoma growth and 2) secondary, which are induced by microglia activation, mitochondrial dysfunction, neurotransmitter and inflammatory mediator release. Although these events typically lead to necrosis, the occurrence of programmed cell death has also been reported after ICH. Methods: We reviewed recent publications describing advance in pre- and clinic ICH research. Results: At present, treatment of ICH patients is based on oral anticoagulant reversal, management of blood pressure and other medical complications. Several pre-clinical studies showed promising results and demonstrated that anti-oxidative and anti-inflammatory treatments reduced neuronal cell death, however, to date, all of these attempts have failed in randomized controlled clinical trials. Yet, the time frame of administration may be crucial in translation from animal to clinical studies. Furthermore, the latest pre-clinical research points toward the existence of other, apoptosis-unrelated forms kinds of pro-grammed cell death. Conclusion: Our review summarizes current knowledge of pathways leading to programmed cell death after ICH in addition to data from clinical trials. Some of the pre-clinical results have not yet demonstrated clinical confirmation, however they sig-nificantly contribute to our understanding of post-ICH pathology and can contribute to development of new therapeutic ap-proaches, decreasing mortality and improving ICH patients’ quality of life.
Collapse
Affiliation(s)
- Tobias Bobinger
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Petra Burkardt
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Hagen B Huttner
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| |
Collapse
|
21
|
Lee RHC, Lee MHH, Wu CYC, Couto e Silva A, Possoit HE, Hsieh TH, Minagar A, Lin HW. Cerebral ischemia and neuroregeneration. Neural Regen Res 2018; 13:373-385. [PMID: 29623912 PMCID: PMC5900490 DOI: 10.4103/1673-5374.228711] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Cerebral ischemia is one of the leading causes of morbidity and mortality worldwide. Although stroke (a form of cerebral ischemia)-related costs are expected to reach 240.67 billion dollars by 2030, options for treatment against cerebral ischemia/stroke are limited. All therapies except anti-thrombolytics (i.e., tissue plasminogen activator) and hypothermia have failed to reduce neuronal injury, neurological deficits, and mortality rates following cerebral ischemia, which suggests that development of novel therapies against stroke/cerebral ischemia are urgently needed. Here, we discuss the possible mechanism(s) underlying cerebral ischemia-induced brain injury, as well as current and future novel therapies (i.e., growth factors, nicotinamide adenine dinucleotide, melatonin, resveratrol, protein kinase C isozymes, pifithrin, hypothermia, fatty acids, sympathoplegic drugs, and stem cells) as it relates to cerebral ischemia.
Collapse
Affiliation(s)
- Reggie H. C. Lee
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Michelle H. H. Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Celeste Y. C. Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Alexandre Couto e Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Harlee E. Possoit
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Tsung-Han Hsieh
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
- Cardiovascular and Metabolomics Research Center, Hualien Tzu Chi Hospital, Hualien, Taiwan, China
| |
Collapse
|
22
|
Park HS, Choi JH. Safety and Efficacy of Hypothermia (34°C) after Hemicraniectomy for Malignant MCA Infarction. J Korean Neurosurg Soc 2018. [PMID: 29526071 PMCID: PMC5853190 DOI: 10.3340/jkns.2016.1111.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE The beneficial effect of hypothermia after hemicraniectomy in malignant middle cerebral artery (MCA) infarction has been controversial. We aim to investigate the safety and clinical efficacy of hypothermia after hemicraniectomy in malignant MCA infarction. METHODS From October 2012 to February 2016, 20 patients underwent hypothermia (Blanketrol III, Cincinnati Sub-Zero, Cincinnati, OH, USA) at 34°C after hemicraniectomy in malignant MCA infarction (hypothermia group). The indication of hypothermia included acute cerebral infarction >2/3 of MCA territory and a Glasgow coma scale (GCS) score <11 with a midline shift >10 mm or transtentorial herniation sign (a fixed and dilated pupil). We retrospectively collected 27 patients, as the control group, who had undergone hemicraniectomy alone and simultaneously met the inclusion criteria of hypothermia between January 2010 and September 2012, before hypothermia was implemented as a treatment strategy in Dong-A University Hospital. We compared the mortality rate between the two groups and investigated hypothermia-related complications, such as postoperative bleeding, pneumonia, sepsis and arrhythmia. RESULTS The age, preoperative infarct volume, GCS score, National institutes of Health Stroke Scale score, and degree of midline shift were not significantly different between the two groups. Of the 20 patients in the hypothermia group, 11 patients were induced with hypothermia immediately after hemicraniectomy and hypothermia was initiated in 9 patients after the decision of hypothermia during postoperative care. The duration of hypothermia was 4±2 days (range, 1 to 7 days). The side effects of hypothermia included two patients with arrhythmia, one with sepsis, one with pneumonia, and one with hypotension. Three cases of hypothermia were discontinued due to these side effects (one sepsis, one hypotension, and one bradycardia). The mortality rate of the hypothermia group was 15.0% and that of the control group was 40.7% (p=0.056). On the basis of the logistic regression analysis, hypothermia was considered to contribute to the decrease in mortality rate (odds ratio, 6.21; 95% confidence interval, 1.04 to 37.05; p=0.045). CONCLUSION This study suggests that hypothermia after hemicraniectomy is a viable option when the progression of patients with malignant MCA infarction indicate poor prognosis.
Collapse
Affiliation(s)
- Hyun-Seok Park
- Department of Neurosurgery, Busan-Ulsan Regional Cardio-Cerebrovascular Center, Medical Science Research Center, Dong-A University College of Medicine, Busan, Korea
| | - Jae-Hyung Choi
- Department of Neurosurgery, Busan-Ulsan Regional Cardio-Cerebrovascular Center, Medical Science Research Center, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
23
|
Abstract
Therapeutic hypothermia (TH) is a potent neuroprotective therapy in experimental cerebral ischemia, with multiple effects at several stages of the ischemic cascade. In animals, TH is so powerful that all preclinical stroke studies require strict temperature control. In humans, multiple clinical studies documented powerful protection with TH after accidental neonatal hypoxic-ischemic injury and global cerebral ischemia with return of spontaneous circulation after cardiac arrest. National and international guidelines recommend TH for selected survivors of global ischemia, with profound benefits seen. Recently, a study comparing target temperature 33-36°C failed to demonstrate significant effects in cardiac arrest patients. Additionally, clinical trials of TH for head trauma and stroke have so far failed to confirm benefit in humans despite a vast preclinical literature. Therefore, it is now critical to understand the fundamental explanation for the success of TH in some, but famously not all, clinical trials. TH in animals appears to work when used soon after ischemia onset; for a short duration; and at a deep target temperature.
Collapse
|
24
|
Sandu RE, Dumbrava D, Surugiu R, Glavan DG, Gresita A, Petcu EB. Cellular and Molecular Mechanisms Underlying Non-Pharmaceutical Ischemic Stroke Therapy in Aged Subjects. Int J Mol Sci 2017; 19:ijms19010099. [PMID: 29286319 PMCID: PMC5796049 DOI: 10.3390/ijms19010099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/22/2017] [Accepted: 12/24/2017] [Indexed: 12/12/2022] Open
Abstract
The incidence of ischemic stroke in humans increases exponentially above 70 years both in men and women. Comorbidities like diabetes, arterial hypertension or co-morbidity factors such as hypercholesterolemia, obesity and body fat distribution as well as fat-rich diet and physical inactivity are common in elderly persons and are associated with higher risk of stroke, increased mortality and disability. Obesity could represent a state of chronic inflammation that can be prevented to some extent by non-pharmaceutical interventions such as calorie restriction and hypothermia. Indeed, recent results suggest that H₂S-induced hypothermia in aged, overweight rats could have a higher probability of success in treating stroke as compared to other monotherapies, by reducing post-stroke brain inflammation. Likewise, it was recently reported that weight reduction prior to stroke, in aged, overweight rats induced by caloric restriction, led to an early re-gain of weight and a significant improvement in recovery of complex sensorimotor skills, cutaneous sensitivity, or spatial memory. CONCLUSION animal models of stroke done in young animals ignore age-associated comorbidities and may explain, at least in part, the unsuccessful bench-to-bedside translation of neuroprotective strategies for ischemic stroke in aged subjects.
Collapse
Affiliation(s)
- Raluca Elena Sandu
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | - Danut Dumbrava
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | - Roxana Surugiu
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | - Daniela-Gabriela Glavan
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | - Andrei Gresita
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | - Eugen Bogdan Petcu
- Gold Coast Campus, School of Medicine, Griffith University, Southport 4222, Australia.
| |
Collapse
|
25
|
Abraini JH, David HN, Blatteau JÉ, Risso JJ, Vallée N. A method for calculating the gas volume proportions and inhalation temperature of inert gas mixtures allowing reaching normothermic or hypothermic target body temperature in the awake rat. Med Gas Res 2017; 7:175-180. [PMID: 29152210 PMCID: PMC5674655 DOI: 10.4103/2045-9912.215746] [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] [Indexed: 11/04/2022] Open
Abstract
The noble gases xenon (Xe) and helium (He) are known to possess neuroprotective properties. Xe is considered the golden standard neuroprotective gas. However, Xe has a higher molecular weight and lower thermal conductivity and specific heat than those of nitrogen, the main diluent of oxygen (O2) in air, conditions that could impair or at least reduce the intrinsic neuroprotective properties of Xe by increasing the critical care patient's respiratory workload and body temperature. In contrast, He has a lower molecular weight and higher thermal conductivity and specific heat than those of nitrogen, but is unfortunately far less potent than Xe at providing neuroprotection. Therefore, combining Xe with He could allow obtaining, depending on the gas inhalation temperature and composition, gas mixtures with neutral or hypothermic properties, the latter being advantageous in term of neuroprotection. However, calculating the thermal properties of a mixture, whatever the substances - gases, metals, rubbers, etc. - is not trivial. To answer this question, we provide a graphical method to assess the volume proportions of Xe, He and O2 that a gas mixture should contain, and the inhalation temperature to which it should be administered to allow a clinician to maintain the patient at a target body temperature.
Collapse
Affiliation(s)
- Jacques H Abraini
- Institut de Recherche Biomédicale des Armées, Équipe Résidente de Recherche Subaquatique Opérationnelle, Toulon, France.,Université Laval, Faculté de Médecine, Département d'Anesthesiologie, Québec, QC, Canada
| | - Hélène N David
- Université Laval, Faculté de Médecine, Département d'Anesthesiologie, Québec, QC, Canada
| | - Jean-Éric Blatteau
- Hôpital d'Instruction des Armées Sainte-Anne, Service de Médecine Hyperbare et Expertise Plongée, Toulon, France
| | - Jean Jacques Risso
- Institut de Recherche Biomédicale des Armées, Équipe Résidente de Recherche Subaquatique Opérationnelle, Toulon, France
| | - Nicolas Vallée
- Institut de Recherche Biomédicale des Armées, Équipe Résidente de Recherche Subaquatique Opérationnelle, Toulon, France
| |
Collapse
|
26
|
Forreider B, Pozivilko D, Kawaji Q, Geng X, Ding Y. Hibernation-like neuroprotection in stroke by attenuating brain metabolic dysfunction. Prog Neurobiol 2017; 157:174-187. [PMID: 26965388 DOI: 10.1016/j.pneurobio.2016.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 11/24/2022]
Abstract
Many mammalian species naturally undergo hibernation, a process that is associated with drastic changes in metabolism and systemic physiology. Their ability to retain an undamaged central nervous system during severely reduced cerebral blood flow has been studied for possible therapeutic application in human ischemic stroke. By inducing a less extreme 'hibernation-like' state, it has been hypothesized that similar neuroprotective effects reduce ischemia-mediated tissue damage in stroke patients. This manuscript includes reviews and evaluations of: (1) true hibernation, (2) hibernation-like state and its neuroprotective characteristics, (3) the preclinical and clinical methods for induction of artificial hibernation (i.e., therapeutic hypothermia, phenothiazine drugs, and ethanol), and (4) the mechanisms by which cerebral ischemia leads to tissue damage and how the above-mentioned induction methods function to inhibit those processes.
Collapse
Affiliation(s)
- Brian Forreider
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - David Pozivilko
- Michigan State University College of Human Medicine, East Lansing, MI, USA
| | - Qingwen Kawaji
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.
| | - Yuchuan Ding
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Dangarembizi R, Erlwanger KH, Mitchell D, Hetem RS, Madziva MT, Harden LM. Measurement of body temperature in normothermic and febrile rats: Limitations of using rectal thermometry. Physiol Behav 2017; 179:162-167. [DOI: 10.1016/j.physbeh.2017.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 01/24/2023]
|
28
|
Rewell SSJ, Jeffreys AL, Sastra SA, Cox SF, Fernandez JA, Aleksoska E, van der Worp HB, Churilov L, Macleod MR, Howells DW. Hypothermia revisited: Impact of ischaemic duration and between experiment variability. J Cereb Blood Flow Metab 2017; 37:3380-3390. [PMID: 28084873 PMCID: PMC5624387 DOI: 10.1177/0271678x16688704] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
To assess the true effect of novel therapies for ischaemic stroke, a positive control that can validate the experimental model and design is vital. Hypothermia may be a good candidate for such a positive control, given the convincing body of evidence from animal models of ischaemic stroke. Taking conditions under which substantial efficacy had been seen in a meta-analysis of hypothermia for focal ischaemia in animal models, we undertook three randomised and blinded studies examining the effect of hypothermia induced immediately following the onset of middle cerebral artery occlusion on infarct volume in rats (n = 15, 23, 264). Hypothermia to a depth of 33℃ and maintained for 130 min significantly reduced infarct volume compared to normothermia treatment (by 27-63%) and depended on ischaemic duration (F(3,244) = 21.242, p < 0.05). However, the protective effect varied across experiments with differences in both the size of the infarct observed in normothermic controls and the time to reach target temperature. Our results highlight the need for sample size and power calculations to take into account variations between individual experiments requiring induction of focal ischaemia.
Collapse
Affiliation(s)
- Sarah SJ Rewell
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Amy L Jeffreys
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Steven A Sastra
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Susan F Cox
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - John A Fernandez
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Elena Aleksoska
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - H Bart van der Worp
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leonid Churilov
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Malcolm R Macleod
- Department of Clinical Neurosciences, University of Edinburgh, Edinburgh, UK
| | - David W Howells
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Heidelberg, Australia
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania
- David W Howells, School of Medicine, Faculty of Health, University of Tasmania, Medical Science Precinct, 17 Liverpool Street, Hobart, Tasmania, Australia.
| |
Collapse
|
29
|
Wu D, Shi J, Elmadhoun O, Duan Y, An H, Zhang J, He X, Meng R, Liu X, Ji X, Ding Y. Dihydrocapsaicin (DHC) enhances the hypothermia-induced neuroprotection following ischemic stroke via PI3K/Akt regulation in rat. Brain Res 2017; 1671:18-25. [PMID: 28684048 DOI: 10.1016/j.brainres.2017.06.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Hypothermia has demonstrated neuroprotection following ischemia in preclinical studies while its clinical application is still very limited. The aim of this study was to explore whether combining local hypothermia in ischemic territory achieved by intra-arterial cold infusions (IACIs) with pharmacologically induced hypothermia enhances therapeutic outcomes, as well as the underlying mechanism. METHODS Sprague-Dawley rats were subjected to right middle cerebral artery occlusion (MCAO) for 2h using intraluminal hollow filament. The ischemic rats were randomized to receive: 1) pharmacological hypothermia by intraperitoneal (i.p.) injection of dihydrocapsaicin (DHC); 2) physical hypothermia by IACIs for 10min; or 3) the combined treatments. Extent of brain injury was determined by neurological deficit, infarct volume, and apoptotic cell death at 24h and/or 7d following reperfusion. ATP and ROS levels were measured. Expression of p-Akt, cleaved Caspase-3, pro-apoptotic (AIF, Bax) and anti-apoptotic proteins (Bcl-2, Bcl-xL) was evaluated at 24h. Finally, PI3K inhibitor was used to determine the effect of p-Akt. RESULTS DHC or IACIs each exhibited hypothermic effect and neuroprotection in rat MCAO models. The combination of pharmacological and physical approaches led to a faster and sustained reduction in brain temperatures and improved ischemia-induced injury than either alone (P<0.01). Furthermore, the combination treatment favorably increased the expression of anti-apoptotic proteins and decreased pro-apoptotic protein levels (P<0.01 or 0.05). This neuroprotective effect was largely blocked by p-Akt inhibition, indicating a potential role of Akt pathway in this mechanism (P<0.01 or 0.05). CONCLUSIONS The combination approach is able to enhance the efficiency of hypothermia and efficacy of hypothermia-induced neuroprotection following ischemic stroke. The findings here move us a step closer towards translating this long recognized TH from bench to bedside.
Collapse
Affiliation(s)
- Di Wu
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China; Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Jingfei Shi
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Omar Elmadhoun
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yunxia Duan
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hong An
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jun Zhang
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoduo He
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Meng
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiangrong Liu
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.
| | - Yuchuan Ding
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
30
|
Modulation by the Noble Gas Helium of Tissue Plasminogen Activator: Effects in a Rat Model of Thromboembolic Stroke. Crit Care Med 2017; 44:e383-9. [PMID: 26646461 DOI: 10.1097/ccm.0000000000001424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTERVENTIONS Helium has been shown to provide neuroprotection in mechanical model of acute ischemic stroke by inducing hypothermia, a condition shown by itself to reduce the thrombolytic and proteolytic properties of tissue plasminogen activator. However, whether or not helium interacts with the thrombolytic drug tissue plasminogen activator, the only approved therapy of acute ischemic stroke still remains unknown. This point is not trivial since previous data have shown the critical importance of the time at which the neuroprotective noble gases xenon and argon should be administered, during or after ischemia, in order not to block tissue plasminogen activator-induced thrombolysis and to obtain neuroprotection and inhibition of tissue plasminogen activator-induced brain hemorrhages. MEASUREMENTS AND MAIN RESULTS We show that helium of 25-75 vol% inhibits in a concentration-dependent fashion the catalytic and thrombolytic activity of tissue plasminogen activator in vitro and ex vivo. In vivo, in rats subjected to thromboembolic brain ischemia, we found that intraischemic helium at 75 vol% inhibits tissue plasminogen activator-induced thrombolysis and subsequent reduction of ischemic brain damage and that postischemic helium at 75 vol% reduces ischemic brain damage and brain hemorrhages. CONCLUSIONS In a clinical perspective for the treatment of acute ischemic stroke, these data suggest that helium 1) should not be administered before or together with tissue plasminogen activator therapy due to the risk of inhibiting the benefit of tissue plasminogen activator-induced thrombolysis; and 2) could be an efficient neuroprotective agent if given after tissue plasminogen activator-induced reperfusion.
Collapse
|
31
|
Hayashi K, Kurose Y. Effects of hypothermia on the mechanical behavior of rabbit femoral arteries. J Mech Behav Biomed Mater 2017; 71:148-155. [PMID: 28297683 DOI: 10.1016/j.jmbbm.2017.03.005] [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: 12/31/2016] [Revised: 02/23/2017] [Accepted: 03/05/2017] [Indexed: 10/20/2022]
Abstract
The need to better understand the effects of non-physiological temperatures on arterial wall behavior is becoming more important because of the increased clinical use of hypothermal and hyperthermal treatments. The present study was performed to examine the effects of temperature on the mechanical behavior of femoral arteries excised from rabbits. Among 17, 27, 37, and 42°C, there were no significant differences in their diameter, stiffness, and P-D relations under the physiologically normal, control condition, although the arterial diameter was slightly smaller at 42°C than at the other three temperatures. Under the SMC-activated condition, on the other hand, we observed significant effects of temperature. For example, arterial diameter at 100mmHg was significantly larger at 17 and 27°C and smaller at 42°C compared with 37°C. Arterial stiffness at 40mmHg were significantly smaller at 17 and larger at 42°C than at 37°C, while the stiffness at 160mmHg were significantly larger at 17°C than at 37°C; however, there were no significant differences in the stiffness at 100mmHg among the four temperatures. Arterial contraction induced by SMC-activation was significantly different between 37°C and the other three temperatures; both of the maximum diameter response and diameter response at 100mmHg were significantly smaller at 17 and 27°C and larger at 42°C compared with 37°C. These results indicate that in the hypothermic range under the control condition, arteries are dilated when cooled, while they are constricted when heated. On the other hand, arterial response to SMC activation is significantly affected by the alterations of temperature. These results indicate that in the hypothermic range under the control condition, arteries are dilated when cooled, while they are constricted when heated. On the other hand, arterial response to the activation of vascular smooth muscle cells is significantly affected by the alteration of temperature. As the mechanical behavior of arterial wall is significantly influenced by temperature, this should be considered in the development of therapeutic methods and techniques for cardiovascular diseases.
Collapse
Affiliation(s)
- Kozaburo Hayashi
- Department of Biomedical Engineering, Okayama University of Science, Okayama 700-0005, Japan.
| | - Yuki Kurose
- Department of Biomedical Engineering, Okayama University of Science, Okayama 700-0005, Japan
| |
Collapse
|
32
|
Ji YB, Zhuang PP, Ji Z, Huang KB, Gu Y, Wu YM, Pan SY, Hu YF. TFP5 is comparable to mild hypothermia in improving neurological outcomes in early-stage ischemic stroke of adult rats. Neuroscience 2017; 343:337-345. [DOI: 10.1016/j.neuroscience.2016.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/19/2016] [Accepted: 12/06/2016] [Indexed: 11/28/2022]
|
33
|
Liu K, Khan H, Geng X, Zhang J, Ding Y. Pharmacological hypothermia: a potential for future stroke therapy? Neurol Res 2017; 38:478-90. [PMID: 27320243 DOI: 10.1080/01616412.2016.1187826] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Mild physical hypothermia after stroke has been associated with positive outcomes. Despite the well-studied beneficial effects of hypothermia in the treatment of stroke, lack of precise temperature control, intolerance for the patient, and immunosuppression are some of the reasons which limit its clinical translation. Pharmacologically induced hypothermia has been explored as a possible treatment option following stroke in animal models. Currently, there are eight classes of pharmacological agents/agonists with hypothermic effects affecting a multitude of systems including cannabinoid, opioid, transient receptor potential vanilloid 1 (TRPV1), neurotensin, thyroxine derivatives, dopamine, gas, and adenosine derivatives. Interestingly, drugs in the TRPV1, neurotensin, and thyroxine families have been shown to have effects in thermoregulatory control in decreasing the compensatory hypothermic response during cooling. This review will briefly present drugs in the eight classes by summarizing their proposed mechanisms of action as well as side effects. Reported thermoregulatory effects of the drugs will also be presented. This review offers the opinion that these agents may be useful in combination therapies with physical hypothermia to achieve faster and more stable temperature control in hypothermia.
Collapse
Affiliation(s)
- Kaiyin Liu
- a Department of Neurological Surgery , Wayne State University School of Medicine , Detroit , MI , USA
| | - Hajra Khan
- a Department of Neurological Surgery , Wayne State University School of Medicine , Detroit , MI , USA
| | - Xiaokun Geng
- a Department of Neurological Surgery , Wayne State University School of Medicine , Detroit , MI , USA.,b Department of Neurology, Beijing Luhe Hospital , Capital Medical University , Beijing , China
| | - Jun Zhang
- c China-America Institute of Neuroscience, Xuanwu Hospital , Capital Medical University , Beijing , China
| | - Yuchuan Ding
- a Department of Neurological Surgery , Wayne State University School of Medicine , Detroit , MI , USA.,b Department of Neurology, Beijing Luhe Hospital , Capital Medical University , Beijing , China
| |
Collapse
|
34
|
Rim HT, Ahn JH, Kim JH, Oh JK, Song JH, Chang IB. Therapeutic Hypothermia for Increased Intracranial Pressure after Decompressive Craniectomy: A Single Center Experience. Korean J Neurotrauma 2016; 12:55-60. [PMID: 27857908 PMCID: PMC5110919 DOI: 10.13004/kjnt.2016.12.2.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/30/2016] [Accepted: 10/12/2016] [Indexed: 01/19/2023] Open
Abstract
Objective Therapeutic hypothermia (TH) and decompressive craniectomy are neuroprotective interventions following severe brain swelling. The precise benefits, risks, and clinical outcomes in brain swelling after TH are still being investigated. We aimed to investigate the effects of TH in severe brain injury after decompressive craniectomy. Methods We reviewed the cases of 24 patients who underwent decompressive craniectomy with intracranial pressure (ICP) monitor insertion in one medical center between January 2012 and May 2016. All patients had an ICP greater than 15 mmHg and a Glasgow Coma Scale score of less than 7 at the time of intervention. TH was induced in half of the patients (n=12) directly after surgery; the remaining 12 patients remained normothermic. The ICP, vital signs, complications, and functional outcomes were reviewed and compared between the patient groups. Results The mean ICP in the TH group was significantly lower than in the normothermia group. Complications during the 3 days after surgery were not different between the groups, with the exception of hypokalemia in the TH group. Mortality in the intensive care unit (ICU) was higher in the normothermia group, but the functional outcomes 3 months after surgery were not different between the TH and normothermia groups. Conclusion TH after decompressive craniectomy was effective for lowering ICP in patients with severe brain swelling. TH also reduced mortality in the ICU, but it had no benefit in functional outcomes.
Collapse
Affiliation(s)
- Hyun Taek Rim
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Jun Hyong Ahn
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Ji Hee Kim
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Jae Keun Oh
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Joon Ho Song
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - In Bok Chang
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| |
Collapse
|
35
|
Vieites-Prado A, Iglesias-Rey R, Fernández-Susavila H, da Silva-Candal A, Rodríguez-Castro E, Gröhn OHJ, Wellmann S, Sobrino T, Castillo J, Campos F. Protective Effects and Magnetic Resonance Imaging Temperature Mapping of Systemic and Focal Hypothermia in Cerebral Ischemia. Stroke 2016; 47:2386-96. [PMID: 27491739 DOI: 10.1161/strokeaha.116.014067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/30/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Hypothermia is potentially the most effective protective therapy for brain ischemia; however, its use is limited because of serious side effects. Although focal hypothermia (FH) has a significantly lower stress profile than systemic hypothermia (SH), its efficacy in ischemia has been poorly studied. We aimed to compare the therapeutic effects of each treatment on various short- and long-term clinically relevant end points. METHODS Sprague-Dawley rats were subjected to transient (45 minutes) occlusion of the middle cerebral artery. One hour after arterial reperfusion, animals were randomly assigned to groups for treatment with SH or FH (target temperature: 32°C) for 4 or 24 hours. Lesion volume, edema, functional recovery, and histological markers of cellular injury were evaluated for 1 month after ischemic injury. Effects of SH and FH on cerebral temperature were also analyzed for the first time by magnetic resonance thermometry, an approach that combines spectroscopy with gradient-echo-based phase mapping. RESULTS Both therapeutic approaches reduced ischemic lesion volume (P<0.001), although a longer FH treatment (24 hours) was required to achieve similar protective effects to those induced by 4 hours of SH. In addition, magnetic resonance thermometry demonstrated that systemic hypothermia reduced whole-brain temperature, whereas FH primarily reduced the temperature of the ischemic region. CONCLUSIONS Focal brain hypothermia requires longer cooling periods to achieve the same protective efficacy as SH. However, FH mainly affects the ischemic region, and therefore represents a promising and nonstressful alternative to SH.
Collapse
Affiliation(s)
- Alba Vieites-Prado
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Ramón Iglesias-Rey
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Héctor Fernández-Susavila
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Andrés da Silva-Candal
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Emilio Rodríguez-Castro
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Olli H J Gröhn
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Sven Wellmann
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Tomás Sobrino
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - José Castillo
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.).
| | - Francisco Campos
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.).
| |
Collapse
|
36
|
Wang B, Wu D, Dornbos III D, Shi J, Ma Y, Zhang M, Liu Y, Chen J, Ding Y, Luo Y, Ji X. Local cerebral hypothermia induced by selective infusion of cold lactated ringer’s: a feasibility study in rhesus monkeys. Neurol Res 2016; 38:545-52. [DOI: 10.1080/01616412.2016.1187827] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
Tahir RA, Pabaney AH. Therapeutic hypothermia and ischemic stroke: A literature review. Surg Neurol Int 2016; 7:S381-6. [PMID: 27313963 PMCID: PMC4901811 DOI: 10.4103/2152-7806.183492] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/18/2016] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Ischemic stroke is the fifth leading cause of death in the US. Clinical techniques aimed at helping to reduce the morbidity associated with stroke have been studied extensively, including therapeutic hypothermia. In this study, the authors review the literature regarding the role of therapeutic hypothermia in ischemic stroke to appreciate the evolution of hypothermia technology over several decades and to critically analyze several early clinical studies to validate its use in ischemic stroke. METHODS A comprehensive literature search was performed using PubMed and Google Scholar databases. Search terms included "hypothermia and ischemic stroke" and "therapeutic hypothermia." A comprehensive search of the current clinical trials using clinicaltrials.gov was conducted using the keywords "stroke and hypothermia" to evaluate early and ongoing clinical trials utilizing hypothermia in ischemic stroke. RESULTS A comprehensive review of the evolution of hypothermia in stroke and the current status of this treatment was performed. Clinical studies were critically analyzed to appreciate their strengths and pitfalls. Ongoing and future registered clinical studies were highlighted and analyzed compared to the reported results of previous trials. CONCLUSION Although hypothermia has been used for various purposes over several decades, its efficacy in the treatment of ischemic stroke is debatable. Several trials have proven its safety and feasibility; however, more robust, randomized clinical trials with large volumes of patients are needed to fully establish its utility in the clinical setting.
Collapse
Affiliation(s)
- Rizwan A Tahir
- Department of Neurological Surgery, Henry Ford Hospital, Detroit, Michigan, USA
| | - Aqueel H Pabaney
- Department of Neurological Surgery, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
38
|
Li X, Ji Z, Gu Y, Hu Y, Huang K, Pan S. Mild hypothermia decreases the total clearance of glibenclamide after low dose administration in rats. Neurosci Lett 2015; 614:55-9. [PMID: 26724224 DOI: 10.1016/j.neulet.2015.12.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 11/02/2015] [Accepted: 12/17/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE Low dose glibenclamide exhibits pleiotropic protective effects in different central nervous system diseases. Previously, we have shown that mild hypothermia enhanced the efficacy of glibenclamide in the cultured cortical neuronal cells. This study aims to evaluate the impact of mild hypothermia on the pharmacokinetics of low dose glibenclamide in rats via its cytochrome P450 2C9 (CYP2C9) metabolic pathway. METHODS Male Sprague-Dawley rats were maintained at 37°C (normothermic group) or cooled to 33°C (hypothermic group). Glibenclamide (33μg/kg) or diclofenac (10mg/kg, a probe drug for assessing the activity of CYP2C9 which involves in glibenclamide and diclofenac metabolism in liver) were intravenously administered at 10min after stabilization of temperature. Plasma samples were collected at 9 different time points. Glibenclamide and diclofenac in sera were separated by liquid chromatography and quantified with tandem mass spectrometry. RESULTS Compared with normothermia, mild hypothermia significantly decreased the total clearance of glibenclamide (16.00±4.1-6.72±2.1mL/min/kg; p<0.01), and there was a non-significant trend in a slightly higher half-life, (1.64±0.34-2.71±1.7h, p=0.157). Area under the plasma concentration versus time curve (AUClast) in the hypothermic group was increased (33.2±11-77.8±18hng/mL, p<0.01). Moreover, mild hypothermia reduced the total clearance of diclofenac (10.33±1.53-7.20±1.66mL/min/kg, p<0.01), indicating that the CYP2C9 activity was compromised in reduced temperature. CONCLUSION Mild hypothermia reduced the total clearance of glibenclamide, probably via mediating the activity of CYP2C9. The impact of hypothermia in clinical application of glibenclamide should be considered.
Collapse
Affiliation(s)
- Xing Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong Ji
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Gu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
39
|
Twenty-four hours hypothermia has temporary efficacy in reducing brain infarction and inflammation in aged rats. Neurobiol Aging 2015; 38:127-140. [PMID: 26827651 DOI: 10.1016/j.neurobiolaging.2015.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/19/2015] [Accepted: 11/11/2015] [Indexed: 11/23/2022]
Abstract
Stroke is a major cause of disability for which no neuroprotective measures are available. Age is the principal nonmodifiable risk factor for this disease. Previously, we reported that exposure to hydrogen sulfide for 48 hours after stroke lowers whole body temperature and confers neuroprotection in aged animals. Because the duration of hypothermia in most clinical trials is between 24 and 48 hours, we questioned whether 24 hours exposure to gaseous hypothermia confers the same neuroprotective efficacy as 48 hours exposure. We found that a shorter exposure to hypothermia transiently reduced both inflammation and infarct size. However, after 1 week, the infarct size became even larger than in controls and after 2 weeks there was no beneficial effect on regenerative processes such as neurogenesis. Behaviorally, hypothermia also had a limited beneficial effect. Finally, after hydrogen sulfide-induced hypothermia, the poststroke aged rats experienced a persistent sleep impairment during their active nocturnal period. Our data suggest that cellular events that are delayed by hypothermia in aged rats may, in the long term, rebound, and diminish the beneficial effects.
Collapse
|
40
|
Cechmanek BK, Tuor UI, Rushforth D, Barber PA. Very Mild Hypothermia (35°C) Postischemia Reduces Infarct Volume and Blood/Brain Barrier Breakdown Following tPA Treatment in the Mouse. Ther Hypothermia Temp Manag 2015; 5:203-8. [PMID: 26075540 DOI: 10.1089/ther.2015.0010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reperfusion therapies for stroke diminish in effectiveness and safety as time to treatment increases. Hypothermia neuroprotection for stroke is established, but its clinical translation has been hampered by uncertainties regarding optimal temperature and complications associated with moderate hypothermia. Also, hypothermia targeting temperatures of 32-33°C is associated with clinical and logistical problems related to induction and adverse side effects. We hypothesized that ischemic damage and tPA-exacerbated blood/brain barrier (BBB) breakdown produced following 30 minutes of middle cerebral artery occlusion and either 1 hour of saline or tPA infusion would be reduced by treatment with very mild cooling of 1.5°C for 48 hours followed by 24 hours of gradual rewarming. Infarct volume was reduced by 29.6% (p<0.001) and 41.9% (p<0.001) in hypothermic-tPA (Hypo_tPA)-treated and hypothermic-saline (Hypo_Sal)-treated animals compared to normothermic-tPA (Norm_tPA) and saline (Norm_Sal)-treated animals, respectively. Hypothermia also reduced IgG extravasation in tPA-treated, but not saline-treated groups compared to their normothermic controls (p<0.001). The ipsilateral-contralateral changes in optical density for IgG extravasation were 18.4% greater in the Norm_tPA than Norm_Sal (p<0.001) group. The ipsilateral-contralateral changes in optical density for IgG extravasation were reduced by 17.8% (p<0.001) in the Hypo_tPA compared to Norm_tPA group. No significant mean difference in IgG extravasation was seen between Hypo_tPA and Hypo_Sal groups (p>0.05). Very modest hypothermia to reduce the BBB breakdown could improve the availability and safety of reperfusion treatments for stroke.
Collapse
Affiliation(s)
- Brian K Cechmanek
- Department of Clinical Neurosciences, Faculty of Medicine, Experimental Imaging Centre and Hotchkiss Brain Institute, University of Calgary , Calgary, Canada
| | - Ursula I Tuor
- Department of Clinical Neurosciences, Faculty of Medicine, Experimental Imaging Centre and Hotchkiss Brain Institute, University of Calgary , Calgary, Canada
| | - David Rushforth
- Department of Clinical Neurosciences, Faculty of Medicine, Experimental Imaging Centre and Hotchkiss Brain Institute, University of Calgary , Calgary, Canada
| | - Philip A Barber
- Department of Clinical Neurosciences, Faculty of Medicine, Experimental Imaging Centre and Hotchkiss Brain Institute, University of Calgary , Calgary, Canada
| |
Collapse
|
41
|
Han Z, Liu X, Luo Y, Ji X. Therapeutic hypothermia for stroke: Where to go? Exp Neurol 2015; 272:67-77. [PMID: 26057949 DOI: 10.1016/j.expneurol.2015.06.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/16/2015] [Accepted: 06/04/2015] [Indexed: 01/08/2023]
Abstract
Ischemic stroke is a major cause of death and long-term disability worldwide. Thrombolysis with recombinant tissue plasminogen activator is the only proven and effective treatment for acute ischemic stroke; however, therapeutic hypothermia is increasingly recognized as having a tissue-protective function and positively influencing neurological outcome, especially in cases of ischemia caused by cardiac arrest or hypoxic-ischemic encephalopathy in newborns. Yet, many aspects of hypothermia as a treatment for ischemic stroke remain unknown. Large-scale studies examining the effects of hypothermia on stroke are currently underway. This review discusses the mechanisms underlying the effect of hypothermia, as well as trends in hypothermia induction methods, methods for achieving optimal protection, side effects, and therapeutic strategies combining hypothermia with other neuroprotective treatments. Finally, outstanding issues that must be addressed before hypothermia treatment is implemented at a clinical level are also presented.
Collapse
Affiliation(s)
- Ziping Han
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xiangrong Liu
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
| | - Xunming Ji
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.
| |
Collapse
|
42
|
Andresen M, Gazmuri JT, Marín A, Regueira T, Rovegno M. Therapeutic hypothermia for acute brain injuries. Scand J Trauma Resusc Emerg Med 2015; 23:42. [PMID: 26043908 PMCID: PMC4456795 DOI: 10.1186/s13049-015-0121-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 04/29/2015] [Indexed: 02/07/2023] Open
Abstract
Therapeutic hypothermia, recently termed target temperature management (TTM), is the cornerstone of neuroprotective strategy. Dating to the pioneer works of Fay, nearly 75 years of basic and clinical evidence support its therapeutic value. Although hypothermia decreases the metabolic rate to restore the supply and demand of O₂, it has other tissue-specific effects, such as decreasing excitotoxicity, limiting inflammation, preventing ATP depletion, reducing free radical production and also intracellular calcium overload to avoid apoptosis. Currently, mild hypothermia (33°C) has become a standard in post-resuscitative care and perinatal asphyxia. However, evidence indicates that hypothermia could be useful in neurologic injuries, such as stroke, subarachnoid hemorrhage and traumatic brain injury. In this review, we discuss the basic and clinical evidence supporting the use of TTM in critical care for acute brain injury that extends beyond care after cardiac arrest, such as for ischemic and hemorrhagic strokes, subarachnoid hemorrhage, and traumatic brain injury. We review the historical perspectives of TTM, provide an overview of the techniques and protocols and the pathophysiologic consequences of hypothermia. In addition, we include our experience of managing patients with acute brain injuries treated using endovascular hypothermia.
Collapse
Affiliation(s)
- Max Andresen
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta, 367, Santiago, Chile.
| | - Jose Tomás Gazmuri
- Hospital de Urgencia Asistencia Pública, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Arnaldo Marín
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta, 367, Santiago, Chile.
| | - Tomas Regueira
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta, 367, Santiago, Chile.
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta, 367, Santiago, Chile.
| |
Collapse
|
43
|
Teismann IK, Oelschläger C, Werstler N, Korsukewitz C, Minnerup J, Ringelstein E, Dziewas R. Discontinuous versus Continuous Weaning in Stroke Patients. Cerebrovasc Dis 2015; 39:269-77. [DOI: 10.1159/000381222] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/17/2015] [Indexed: 11/19/2022] Open
Abstract
Background: An increasing number of stroke patients have to be supported by mechanical ventilation in intensive care units (ICU), with a relevant proportion of them requiring gradual withdrawal from a respirator. To date, weaning studies have focused merely on mixed patient groups, COPD patients or patients after cardiac surgery. Therefore, the best weaning strategy for stroke patients remains to be determined. Methods: Here, we designed a prospective randomized controlled study comparing adaptive support ventilation (ASV), a continuous weaning strategy, with biphasic positive airway pressure (BIPAP) in combination with spontaneous breathing trials, a discontinuous technique, in the treatment of stroke patients. The primary endpoint was the duration of the weaning process. Results: Only the 40 (out of 54) patients failing in an initial spontaneous breathing trial (T-piece test) were included into the study; the failure proportion is considerably larger compared to previous studies. Eligible patients were pseudo-randomly assigned to one of the two weaning groups. Both groups did not differ regarding age, gender, and severity of stroke. The results showed that the median weaning duration was 10.7 days (±SD 7.0) in the discontinuous weaning group, and 8 days (±SD 4.5) in the continuous weaning group (p < 0.05). Conclusions: To the best of our knowledge, this is the first clinical study to show that continuous weaning is significantly more effective compared to discontinuous weaning in mechanically ventilated stroke patients. We suppose that the reason for the superiority of continuous weaning using ASV as well as the bad performance of our patients in the 2 h T-piece test is caused by the patients' compliance. Compared to patients on surgical and medical ICUs, neurological patients more often suffer from reduced vigilance, lack of adverse-effects reflexes, dysphagia, and cerebral dysfunction. Therefore, stroke patients may profit from a more gradual withdrawal of weaning.
Collapse
|
44
|
Esposito E, Ebner M, Ziemann U, Poli S. In cold blood: intraarteral cold infusions for selective brain cooling in stroke. J Cereb Blood Flow Metab 2014; 34:743-52. [PMID: 24517972 PMCID: PMC4013766 DOI: 10.1038/jcbfm.2014.29] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/19/2013] [Accepted: 01/19/2014] [Indexed: 12/29/2022]
Abstract
Hypothermia is a promising therapeutic option for stroke patients and an established neuroprotective treatment for global cerebral ischemia after cardiac arrest. While whole body cooling is a feasible approach in intubated and sedated patients, its application in awake stroke patients is limited by severe side effects: Strong shivering rewarms the body and potentially worsens ischemic conditions because of increased O2 consumption. Drugs used for shivering control frequently cause sedation that increases the risk of aspiration and pneumonia. Selective brain cooling by intraarterial cold infusions (IACIs) has been proposed as an alternative strategy for patients suffering from acute ischemic stroke. Preclinical studies and early clinical experience indicate that IACI induce a highly selective brain temperature decrease within minutes and reach targeted hypothermia 10 to 30 times faster than conventional cooling methods. At the same time, body core temperature remains largely unaffected, thus systemic side effects are potentially diminished. This review critically discusses the limitations and side effects of current cooling techniques for neuroprotection from ischemic brain damage and summarizes the available evidence regarding advantages and potential risks of IACI.
Collapse
Affiliation(s)
- Elga Esposito
- Department Neurology & Stroke, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Ebner
- Department Neurology & Stroke, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ulf Ziemann
- Department Neurology & Stroke, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Sven Poli
- Department Neurology & Stroke, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
45
|
Varendi K, Airavaara M, Anttila J, Vose S, Planken A, Saarma M, Mitchell JR, Andressoo JO. Short-term preoperative dietary restriction is neuroprotective in a rat focal stroke model. PLoS One 2014; 9:e93911. [PMID: 24705386 PMCID: PMC3976327 DOI: 10.1371/journal.pone.0093911] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/07/2014] [Indexed: 01/13/2023] Open
Abstract
Stroke is a major complication of cardiovascular surgery, resulting in over 100,000 deaths and over a million postoperative encephalopathies annually in the US and Europe. While mitigating damage from stroke after it occurs has proven elusive, opportunities to reduce the incidence and/or severity of stroke prior to surgery in at-risk individuals remain largely unexplored. We tested the potential of short-term preoperative dietary restriction to provide neuroprotection in rat models of focal stroke. Rats were preconditioned with either three days of water-only fasting or six days of a protein free diet prior to induction of transient middle cerebral artery occlusion using two different methods, resulting in either a severe focal stroke to forebrain and midbrain, or a mild focal stroke localized to cortex only. Infarct volume, functional recovery and molecular markers of damage and protection were assessed up to two weeks after reperfusion. Preoperative fasting for 3 days reduced infarct volume after severe focal stroke. Neuroprotection was associated with modulation of innate immunity, including elevation of circulating neutrophil chemoattractant C-X-C motif ligand 1 prior to ischemia and suppression of striatal pro-inflammatory markers including tumor necrosis factor α, its receptor and downstream effector intercellular adhesion molecule-1 after reperfusion. Similarly, preoperative dietary protein restriction for 6 days reduced ischemic injury and improved functional recovery in a milder cortical infarction model. Our results suggest that short-term dietary restriction regimens may provide simple and translatable approaches to reduce perioperative stroke severity in high-risk elective vascular surgery.
Collapse
Affiliation(s)
- Kärt Varendi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jenni Anttila
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Sarah Vose
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Anu Planken
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
- * E-mail: (JRM); (JOA)
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- * E-mail: (JRM); (JOA)
| |
Collapse
|
46
|
|
47
|
Ji Y, Hu Y, Wu Y, Ji Z, Song W, Wang S, Pan S. Therapeutic time window of hypothermia is broader than cerebral artery flushing in carotid saline infusion after transient focal ischemic stroke in rats. Neurol Res 2013; 34:657-63. [PMID: 22709718 DOI: 10.1179/1743132812y.0000000061] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Yabin Ji
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongming Wu
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong Ji
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Song
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengnan Wang
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of NeurologyNanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
48
|
Murtha LA, McLeod DD, McCann SK, Pepperall D, Chung S, Levi CR, Calford MB, Spratt NJ. Short-duration hypothermia after ischemic stroke prevents delayed intracranial pressure rise. Int J Stroke 2013; 9:553-9. [PMID: 24025084 DOI: 10.1111/ijs.12181] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/22/2013] [Indexed: 12/01/2022]
Abstract
BACKGROUND Intracranial pressure elevation, peaking three to seven post-stroke is well recognized following large strokes. Data following small-moderate stroke are limited. Therapeutic hypothermia improves outcome after cardiac arrest, is strongly neuroprotective in experimental stroke, and is under clinical trial in stroke. Hypothermia lowers elevated intracranial pressure; however, rebound intracranial pressure elevation and neurological deterioration may occur during rewarming. HYPOTHESES (1) Intracranial pressure increases 24 h after moderate and small strokes. (2) Short-duration hypothermia-rewarming, instituted before intracranial pressure elevation, prevents this 24 h intracranial pressure elevation. METHODS Long-Evans rats with two hour middle cerebral artery occlusion or outbred Wistar rats with three hour middle cerebral artery occlusion had intracranial pressure measured at baseline and 24 h. Wistars were randomized to 2·5 h hypothermia (32·5°C) or normothermia, commencing 1 h after stroke. RESULTS In Long-Evans rats (n = 5), intracranial pressure increased from 10·9 ± 4·6 mmHg at baseline to 32·4 ± 11·4 mmHg at 24 h, infarct volume was 84·3 ± 15·9 mm(3) . In normothermic Wistars (n = 10), intracranial pressure increased from 6·7 ± 2·3 mmHg to 31·6 ± 9·3 mmHg, infarct volume was 31·3 ± 18·4 mm(3) . In hypothermia-treated Wistars (n = 10), 24 h intracranial pressure did not increase (7·0 ± 2·8 mmHg, P < 0·001 vs. normothermia), and infarct volume was smaller (15·4 ± 11·8 mm(3) , P < 0·05). CONCLUSIONS We saw major intracranial pressure elevation 24 h after stroke in two rat strains, even after small strokes. Short-duration hypothermia prevented the intracranial pressure rise, an effect sustained for at least 18 h after rewarming. The findings have potentially important implications for design of future clinical trials.
Collapse
Affiliation(s)
- L A Murtha
- University of Newcastle and Hunter Medical Research Institute, School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Altınsoy C, Tuzun F, Duman N, Sever AH, Dilek M, Ozbal S, Ergur BU, Yesilirmak DC, Yılmaz O, Kumral A, Ozkan H. Effect of induced hypothermia on lipopolysaccharide-induced lung injury in neonatal rats. J Matern Fetal Neonatal Med 2013; 27:421-9. [DOI: 10.3109/14767058.2013.818115] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
50
|
Wei S, Sun J, Li J, Wang L, Hall CL, Dix TA, Mohamad O, Wei L, Yu SP. Acute and delayed protective effects of pharmacologically induced hypothermia in an intracerebral hemorrhage stroke model of mice. Neuroscience 2013; 252:489-500. [PMID: 23912033 DOI: 10.1016/j.neuroscience.2013.07.052] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 12/21/2022]
Abstract
Hemorrhagic stroke, including intracerebral hemorrhage (ICH), is a devastating subtype of stroke; yet, effective clinical treatment is very limited. Accumulating evidence has shown that mild to moderate hypothermia is a promising intervention for ischemic stroke and ICH. Current physical cooling methods, however, are less efficient and often impractical for acute ICH patients. The present investigation tested pharmacologically induced hypothermia (PIH) using the second-generation neurotensin receptor (NTR) agonist HPI-201 (formerly known as ABS-201) in an adult mouse model with ICH. Acute or delayed administrations of HPI-201 (2mg/kg bolus injection followed by 2 injections of 1mg/kg, i.p.) were initiated at 1 or 24h after ICH. HPI-201 induced mild hypothermia within 30 min and body and brain temperatures were maintained at 32.7 ± 0.4°C for at least 6h without causing observable shivering. With the 1-h delayed treatment, HPI-201-induced PIH significantly reduced ICH-induced cell death and brain edema compared to saline-treated ICH animals. When HPI-201-induced hypothermia was initiated 24h after the onset of ICH, it still significantly attenuated brain edema, cell death and blood-brain barrier breakdown. HPI-201 significantly decreased the expression of matrix metallopeptidase-9 (MMP-9), reduced caspase-3 activation, and increased Bcl-2 expression in the ICH brain. Moreover, ICH mice received 1-h delayed HPI-201 treatment performed significantly better in the neurological behavior test 48 h after ICH. All together, these data suggest that systemic injection of HPI-201 is an effective hypothermic strategy that protects the brain from ICH injury with a wide therapeutic window. The protective effect of this PIH therapy is partially mediated through the alleviation of apoptosis and neurovascular damage. We suggest that pharmacological hypothermia using the newly developed neurotensin analogs is a promising therapeutic treatment for ICH.
Collapse
Affiliation(s)
- S Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | | | | | | | | | | | | | | | | |
Collapse
|