1
|
Kobata H. Clinical Insights and Future Directions in Hypothermia for Severe Traumatic Brain Injury: A Narrative Review. J Clin Med 2024; 13:4221. [PMID: 39064261 PMCID: PMC11278030 DOI: 10.3390/jcm13144221] [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: 04/12/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Fever control is essential in patients with severe traumatic brain injury (TBI). The efficacy of therapeutic hypothermia (TH) in severe TBI has been investigated over the last few decades; however, in contrast to experimental studies showing benefits, no evidence of efficacy has been demonstrated in clinical practice. In this review, the mechanisms and history of hypothermia were briefly outlined, while the results of major randomized controlled trials (RCTs) and meta-analyses investigating TH for adult TBI were introduced and discussed. The retrieved meta-analyses showed conflicting results, with a limited number of studies indicating the benefits of TH. Some studies have shown the benefits of long-term TH compared with short-term TH. Although TH is effective at lowering elevated intracranial pressure (ICP), reduced ICP does not lead to favorable outcomes. Low-quality RCTs overestimated the benefits of TH, while high-quality RCTs showed no difference or worse outcomes with TH. RCTs assessing standardized TH quality demonstrated the benefits of TH. As TBI has heterogeneous and complicated pathologies, applying a uniform treatment may not be ideal. A meta-analysis of young patients who underwent early cooling and hematoma removal showed better TH results. TH should not be abandoned, and its optimal usage should be advocated on an individual basis.
Collapse
Affiliation(s)
- Hitoshi Kobata
- Department of Emergency and Critical Care Medicine/Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| |
Collapse
|
2
|
Doshi M, Natori Y, Ishii A, Saigusa D, Watanabe S, Hosoyamada M, Hirashima-Akae Y. Hypothermia increases adenosine monophosphate and xanthosine monophosphate levels in the mouse hippocampus, preventing their reduction by global cerebral ischemia. Sci Rep 2024; 14:3187. [PMID: 38326353 PMCID: PMC10850059 DOI: 10.1038/s41598-024-53530-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Global cerebral ischemia (GCI) caused by clinical conditions such as cardiac arrest leads to delayed neuronal death in the hippocampus, resulting in physical and mental disability. However, the mechanism of delayed neuronal death following GCI remains unclear. To elucidate the mechanism, we performed a metabolome analysis using a mouse model in which hypothermia (HT) during GCI, which was induced by the transient occlusion of the bilateral common carotid arteries, markedly suppressed the development of delayed neuronal death in the hippocampus after reperfusion. Fifteen metabolites whose levels were significantly changed by GCI and 12 metabolites whose levels were significantly changed by HT were identified. Furthermore, the metabolites common for both changes were narrowed down to two, adenosine monophosphate (AMP) and xanthosine monophosphate (XMP). The levels of both AMP and XMP were found to be decreased by GCI, but increased by HT, thereby preventing their decrease. In contrast, the levels of adenosine, inosine, hypoxanthine, xanthine, and guanosine, the downstream metabolites of AMP and XMP, were increased by GCI, but were not affected by HT. Our results may provide a clue to understanding the mechanism by which HT during GCI suppresses the development of delayed neuronal death in the hippocampus.
Collapse
Affiliation(s)
- Masaru Doshi
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| | - Yujin Natori
- Department of Legal Medicine and Bioethics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Akira Ishii
- Department of Legal Medicine and Bioethics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Daisuke Saigusa
- Department of Biomedical and Analytical Sciences, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Shiro Watanabe
- Division of Nutritional Biochemistry, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Makoto Hosoyamada
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | | |
Collapse
|
3
|
Sun G, Qin W, Wang Q, Sun X, Chen H, Li J, Sun L, Shi F, Zhang G, Wang M. Selective-cerebral-hypothermia-induced neuroprotection against-focal cerebral ischemia/reperfusion injury is associated with an increase in SUMO2/3 conjugation. Brain Res 2021; 1756:147311. [PMID: 33539797 DOI: 10.1016/j.brainres.2021.147311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 10/22/2022]
Abstract
Selective cerebral hypothermia is considered an effective treatment for neuronal injury after stroke and avoids the complications of general hypothermia. Several recent studies hanve suggested that SUMO2/3 conjugation occurs following cerebral ischemia/reperfusion (I/R) injury. However, the relationship between the cerebral protective effect of selective cerebral hypothermia and SUMO2/3 conjugation remains unclear. In this study, we investigated the effect of selective cerebral hypothermia on SUMO2/3 conjugation during focal cerebral I/R injury. A total of 140 Sprague-Dawley rats were divided into four groups. In the sham group, only the carotid artery was exposed. The endoluminal filament technique was used to induce middle cerebral artery occlusion in the other three groups. After 2 h of occlusion, the filaments were slowly removed to allow blood reperfusion in the I/R group. In the hypothermia (HT) group and normothermia (NT) group, normal saline at 4 °C and 37 °C, respectively , was perfused through the carotid artery, followed by the restoration of blood flow. The results of the modified neurological severity score (mNSS), 2,3,5-triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (HE) staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining demonstrated that selective cerebral hypothermia significantly decreased I/R-induced neuronal injury (mNSS, n = 8, 24 h, HT (5.88 ± 2.36) vs. I/R (8.63 ± 3.38), P < 0.05. 48 h, HT (5.75 ± 2.25) vs. I/R (8.5 ± 2.88), P < 0.05. Cerebral infarct volume percentages, n = 5, HT (18.71 ± 2.13) vs. I/R (41.52 ± 2.90), P < 0.01. Cell apoptosis rate, n = 5, 24 h, HT (21.28 ± 2.61) vs. I/R (43.72 ± 4.30), P < 0.05. 48 h, HT (20.50 ± 2.53) vs. I/R (38.94 ± 2.93), P < 0.05). The expression of Ubc9 and conjugated SUMO2/3 proteins was increased at 24 and 48 h after reperfusion in the 3 non-sham groups, and hypothermia further upregulated the expression of Ubc9 and conjugated SUMO2/3 proteins in the HT group. The expression of SENP3 was increased in the NT group and I/R group, while it was decreased in the HT group at 24 and 48 h after reperfusion (Relative quantities, n = 5, Ubc9, 24 h, HT (2.44 ± 0.22) vs. I/R (1.55 ± 0.39), P < 0.05. 48 h, HT (2.69 ± 0.16) vs. I/R (2.25 ± 0.33), P < 0.05. SENP3, 24 h, HT (0.47 ± 0.15) vs. I/R (2.18 ± 0.43), P < 0.05. 48 h, HT (0.72 ± 0.06) vs. I/R (1.51 ± 0.19), P < 0.05. conjugated SUMO2/3 proteins, 24 h, HT (2.84 ± 0.24) vs. I/R (2.51 ± 0.20), P < 0.05. 48 h, HT (2.73 ± 0.13) vs. I/R (2.44 ± 0.13), P < 0.05). Further analysis showed that the variation in SENP3 expression was more obvious than that in Ubc9 under hypothermia intervention in the HT group. These findings suggest that selective cerebral hypothermia could increase SUMO2/3 modification mainly via down-regulating the expression of SENP3, and then exert neuroprotective effects in rats with cerebral I/R injury.
Collapse
Affiliation(s)
- Guiliang Sun
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Weiwei Qin
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Qiang Wang
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Xiaopeng Sun
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Huailong Chen
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Jingzhu Li
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Lixin Sun
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Fei Shi
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China
| | - Gaofeng Zhang
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China.
| | - Mingshan Wang
- Department of Anesthesiology, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
4
|
Karlsson J, Petersén A, Gidö G, Wieloch T, Brundin P. Combining Neuroprotective Treatment of Embryonic Nigral Donor Tissue with Mild Hypothermia of the Graft Recipient. Cell Transplant 2017; 14:301-9. [PMID: 16052911 DOI: 10.3727/000000005783983089] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Around 80–95% of the immature dopaminergic neurons die when embryonic ventral mesencephalic tissue is transplanted. Cell death occurs both during the preparation of donor tissue and after graft implantation, but the effect of combining successful neuroprotective treatments before and after transplantation has not been extensively investigated. We therefore treated embryonic rat mesencephalic tissue with a combination of the lipid peroxidation inhibitor tirilazad mesylate (3 μM) and the caspase inhibitor Ac.YVAD.cmk (500 μM) and transplanted the tissue into hemiparkinsonian rats kept hypothermic (32–33°C) or normothermic (37°C) during, and 90 min following, graft surgery. Suspension cell number did not differ between untreated or tirilazad/YVAD-treated preparations prior to transplantation. When graft survival was evaluated 6 weeks after implantation, both tirilazad/YVAD pretreatment and mild hypothermia increased the survival of transplanted dopaminergic neurons. Approximately 50–57% of the embryonic dopaminergic neurons survived the dissociation and grafting procedure in rats rendered hypothermic, but there was no significant additive effect on graft survival with a combined treatment. All groups of rats exhibited behavioral recovery in the amphetamine-induced rotation test. There was a significantly enhanced functional capacity of grafts placed in hypothermic as compared to normothermic rats. However, tirilazad/YVAD pretreated implants did not afford greater behavioral improvement than control-treated grafts. Our results suggest that neuroprotective treatments administered prior to and immediately after neural graft implantation may under certain conditions rescue, at least in part, the same subset of dopaminergic neurons. The study also emphasizes the importance of the immediate time after grafting for transplant survival, with relevance both for primary mesencephalic implants and stem cell grafts.
Collapse
Affiliation(s)
- Jenny Karlsson
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, SE-221 84 Lund, Sweden
| | | | | | | | | |
Collapse
|
5
|
Mulder M, Geocadin RG. Neurology of cardiopulmonary resuscitation. HANDBOOK OF CLINICAL NEUROLOGY 2017; 141:593-617. [PMID: 28190437 DOI: 10.1016/b978-0-444-63599-0.00032-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This chapter aims to provide an up-to-date review of the science and clinical practice pertaining to neurologic injury after successful cardiopulmonary resuscitation. The past two decades have seen a major shift in the science and practice of cardiopulmonary resuscitation, with a major emphasis on postresuscitation neurologic care. This chapter provides a nuanced and thoughtful historic and bench-to-bedside overview of the neurologic aspects of cardiopulmonary resuscitation. A particular emphasis is made on the anatomy and pathophysiology of hypoxic-ischemic encephalopathy, up-to-date management of survivors of cardiopulmonary resuscitation, and a careful discussion on neurologic outcome prediction. Guidance to practice evidence-based clinical care when able and thoughtful, pragmatic suggestions for care where evidence is lacking are also provided. This chapter serves as both a useful clinical guide and an updated, thorough, and state-of-the-art reference on the topic for advanced students and experienced practitioners in the field.
Collapse
Affiliation(s)
- M Mulder
- Department of Critical Care and the John Nasseff Neuroscience Institute, Abbott Northwestern Hospital, Allina Health, Minneapolis, MN, USA
| | - R G Geocadin
- Neurosciences Critical Care Division, Department of Anesthesiology and Critical Care Medicine and Departments of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
Zhao DA, Bi LY, Huang Q, Zhang FM, Han ZM. [Isoflurane provides neuroprotection in neonatal hypoxic ischemic brain injury by suppressing apoptosis]. Rev Bras Anestesiol 2016; 66:613-621. [PMID: 27637994 DOI: 10.1016/j.bjan.2016.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/22/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Isoflurane is halogenated volatile ether used for inhalational anesthesia. It is widely used in clinics as an inhalational anesthetic. Neonatal hypoxic ischemia injury ensues in the immature brain that results in delayed cell death via excitotoxicity and oxidative stress. Isoflurane has shown neuroprotective properties that make a beneficial basis of using isoflurane in both cell culture and animal models, including various models of brain injury. We aimed to determine the neuroprotective effect of isoflurane on hypoxic brain injury and elucidated the underlying mechanism. METHODS A hippocampal slice, in artificial cerebrospinal fluid with glucose and oxygen deprivation, was used as an in vitro model for brain hypoxia. The orthodromic population spike and hypoxic injury potential were recorded in the CA1 and CA3 regions. Amino acid neurotransmitters concentration in perfusion solution of hippocampal slices was measured. RESULTS Isoflurane treatment caused delayed elimination of population spike and improved the recovery of population spike; decreased frequency of hypoxic injury potential, postponed the onset of hypoxic injury potential and increased the duration of hypoxic injury potential. Isoflurane treatment also decreased the hypoxia-induced release of amino acid neurotransmitters such as aspartate, glutamate and glycine induced by hypoxia, but the levels of γ-aminobutyric acid were elevated. Morphological studies showed that isoflurane treatment attenuated edema of pyramid neurons in the CA1 region. It also reduced apoptosis as evident by lowered expression of caspase-3 and PARP genes. CONCLUSIONS Isoflurane showed a neuro-protective effect on hippocampal neuron injury induced by hypoxia through suppression of apoptosis.
Collapse
Affiliation(s)
- De-An Zhao
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China.
| | - Ling-Yun Bi
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| | - Qian Huang
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| | - Fang-Min Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| | - Zi-Ming Han
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| |
Collapse
|
7
|
Kim ES, Lee SK, Kwon MJ, Lee PH, Ju YS, Yoon DY, Kim HJ, Lee KS. Assessment of Blood-Brain Barrier Permeability by Dynamic Contrast-Enhanced MRI in Transient Middle Cerebral Artery Occlusion Model after Localized Brain Cooling in Rats. Korean J Radiol 2016; 17:715-24. [PMID: 27587960 PMCID: PMC5007398 DOI: 10.3348/kjr.2016.17.5.715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/17/2016] [Indexed: 12/21/2022] Open
Abstract
Objective The purpose of this study was to evaluate the effects of localized brain cooling on blood-brain barrier (BBB) permeability following transient middle cerebral artery occlusion (tMCAO) in rats, by using dynamic contrast-enhanced (DCE)-MRI. Materials and Methods Thirty rats were divided into 3 groups of 10 rats each: control group, localized cold-saline (20℃) infusion group, and localized warm-saline (37℃) infusion group. The left middle cerebral artery (MCA) was occluded for 1 hour in anesthetized rats, followed by 3 hours of reperfusion. In the localized saline infusion group, 6 mL of cold or warm saline was infused through the hollow filament for 10 minutes after MCA occlusion. DCE-MRI investigations were performed after 3 hours and 24 hours of reperfusion. Pharmacokinetic parameters of the extended Tofts-Kety model were calculated for each DCE-MRI. In addition, rotarod testing was performed before tMCAO, and on days 1-9 after tMCAO. Myeloperoxidase (MPO) immunohisto-chemistry was performed to identify infiltrating neutrophils associated with the inflammatory response in the rat brain. Results Permeability parameters showed no statistical significance between cold and warm saline infusion groups after 3-hour reperfusion 0.09 ± 0.01 min-1 vs. 0.07 ± 0.02 min-1, p = 0.661 for Ktrans; 0.30 ± 0.05 min-1 vs. 0.37 ± 0.11 min-1, p = 0.394 for kep, respectively. Behavioral testing revealed no significant difference among the three groups. However, the percentage of MPO-positive cells in the cold-saline group was significantly lower than those in the control and warm-saline groups (p < 0.05). Conclusion Localized brain cooling (20℃) does not confer a benefit to inhibit the increase in BBB permeability that follows transient cerebral ischemia and reperfusion in an animal model, as compared with localized warm-saline (37℃) infusion group.
Collapse
Affiliation(s)
- Eun Soo Kim
- Department of Radiology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
| | - Seung-Koo Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
| | - Phil Hye Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Young-Su Ju
- Department of Industrial Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
| | - Dae Young Yoon
- Department of Radiology, Hallym University Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 05355, Korea
| | - Hye Jeong Kim
- Department of Radiology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Korea
| | - Kwan Seop Lee
- Department of Radiology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
| |
Collapse
|
8
|
Zhao DA, Bi LY, Huang Q, Zhang FM, Han ZM. Isoflurane provides neuroprotection in neonatal hypoxic ischemic brain injury by suppressing apoptosis. Braz J Anesthesiol 2016; 66:613-621. [PMID: 27793236 DOI: 10.1016/j.bjane.2015.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/22/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Isoflurane is halogenated volatile ether used for inhalational anesthesia. It is widely used in clinics as an inhalational anesthetic. Neonatal hypoxic ischemia injury ensues in the immature brain that results in delayed cell death via excitotoxicity and oxidative stress. Isoflurane has shown neuroprotective properties that make a beneficial basis of using isoflurane in both cell culture and animal models, including various models of brain injury. We aimed to determine the neuroprotective effect of isoflurane on hypoxic brain injury and elucidated the underlying mechanism. METHODS A hippocampal slice, in artificial cerebrospinal fluid with glucose and oxygen deprivation, was used as an in vitro model for brain hypoxia. The orthodromic population spike and hypoxic injury potential were recorded in the CA1 and CA3 regions. Amino acid neurotransmitters concentration in perfusion solution of hippocampal slices was measured. RESULTS Isoflurane treatment caused delayed elimination of population spike and improved the recovery of population spike; decreased frequency of hypoxic injury potential, postponed the onset of hypoxic injury potential and increased the duration of hypoxic injury potential. Isoflurane treatment also decreased the hypoxia-induced release of amino acid neurotransmitters such as aspartate, glutamate and glycine induced by hypoxia, but the levels of γ-aminobutyric acid were elevated. Morphological studies showed that isoflurane treatment attenuated edema of pyramid neurons in the CA1 region. It also reduced apoptosis as evident by lowered expression of caspase-3 and PARP genes. CONCLUSIONS Isoflurane showed a neuro-protective effect on hippocampal neuron injury induced by hypoxia through suppression of apoptosis.
Collapse
Affiliation(s)
- De-An Zhao
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China.
| | - Ling-Yun Bi
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| | - Qian Huang
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| | - Fang-Min Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| | - Zi-Ming Han
- The First Affiliated Hospital of Xinxiang Medical University, Department of Pediatrics, Weihui, China
| |
Collapse
|
9
|
Zhang DX, Ding HZ, Jiang S, Zeng YM, Tang QF. Anin vitrostudy of the neuroprotective effect of propofol on hypoxic hippocampal slice. Brain Inj 2014; 28:1758-65. [DOI: 10.3109/02699052.2014.947624] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
10
|
Sakurai T, Kudo M, Watanabe T, Itoh K, Higashitsuji H, Arizumi T, Inoue T, Hagiwara S, Ueshima K, Nishida N, Fukumoto M, Fujita J. Hypothermia protects against fulminant hepatitis in mice by reducing reactive oxygen species production. Dig Dis 2013; 31:440-6. [PMID: 24281018 DOI: 10.1159/000355242] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Mild hypothermia (32-33°C) shows protective effects in patients with brain damage and cardiac arrest. Although cold-inducible RNA-binding protein (CIRP) contributes to the protective effects of hypothermia through extracellular signal-regulated kinase activation in fibroblasts, the effects of hypothermia in the liver remain unclear. METHODS We analysed the effects of cold temperature on fulminant hepatitis, a potentially fatal disease, using the D-galactosamine (GalN)/lipopolysaccharide (LPS) and concanavalin (con) A-induced hepatitis models in mice. After GalN/LPS administration and anaesthesia, mice in the hypothermia group were kept at 25°C and those in control group were kept at 35°C. After concanavalin A (con A) administration, the mice in the hypothermia group were placed in a chamber with an ambient temperature of 6°C for 1.5 h. RESULTS Hypothermia attenuated liver injury and prolonged survival. Activation of c-Jun N-terminal kinase and Akt, which are involved in reactive oxygen species (ROS) accumulation, was suppressed by low temperature. Hypothermia significantly decreased oxidized protein levels, and treatment with N-acetyl-L-cysteine, an antioxidant, attenuated GalN/LPS-induced liver injury. In con A-induced hepatitis, CIRP expression was upregulated and Bid expression was downregulated, resulting in decreased apoptosis of hepatocytes in the hypothermia group. CONCLUSIONS These data suggest that hypothermia directly protects hepatocytes from cell death via reduction of ROS production in fulminant hepatitis.
Collapse
Affiliation(s)
- Toshiharu Sakurai
- Department of Gastroenterology and Hepatology, Kinki University School of Medicine, Osakasayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
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
|
12
|
Cheng H, Ji X, Ding Y, Luo Y, Wang G, Sun X, Chen J, Ling F. Focal perfusion of circulating cooled blood reduces the infarction volume and improves neurological outcome in middle cerebral artery occlusion. Neurol Res 2013; 31:340-5. [DOI: 10.1179/174313209x443982] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
13
|
Li J, Luan X, Lai Q, Clark JC, McAllister JP, Fessler R, Diaz FG, Ding Y. Long-term neuroprotection induced by regional brain cooling with saline infusion into ischemic territory in rats: a behavioral analysis. Neurol Res 2013; 26:677-83. [PMID: 15327759 DOI: 10.1179/016164104225015903] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The neuroprotective effect of hypothermia has long been recognized. Our recent studies have demonstrated the significant therapeutic value of local brain cooling in the ischemic territory prior to reperfusion in stroke, with reduced infarction and inflammatory responses up to 48 hours of reperfusion. The goal of this study was to determine if local brain cooling, produced by infusion of cold saline, could induce long-term functional improvement after stroke. A hollow filament was used to block the middle cerebral artery (MCA) for 3 hours, and then to locally infuse the ischemic territory with 6 ml cold saline (20 degrees C) for 10 minutes prior to reperfusion. This brain cooling infusion induced a significant (p < 0.01) decrease in neurologic deficits and significantly (p < 0.01) improved motor behavior in ischemic rats after 14 days of reperfusion, compared with ischemic rats without local cold saline infusion. This improvement continued for up to 28 days after reperfusion. No significant difference in motor performance was observed between the brain cooling infusion and normal control groups. Significant (p < 0.01) reductions in infarct volume were also evident. In conclusion, a local cerebral hypothermia induced by local saline infusion prior to reperfusion produced a long-term functional recovery after ischemic stroke. A therapeutic procedure, which combines prereperfusion infusion into an ischemic region with coincident cerebral hypothermia and perhaps subsequent recanalization of an occluded intracranial vessel, may improve the outcome for stroke patients.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurological Surgery, Wayne State University School of Medicine, Canfield, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang F, Luo Y, Ling F, Wu H, Chen J, Yan F, He Z, Goel G, Ji X, Ding Y. Comparison of neuroprotective effects in ischemic rats with different hypothermia procedures. Neurol Res 2013; 32:378-83. [DOI: 10.1179/016164110x12670144526183] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
15
|
Dickman CA. Thoracoscopic correction and placement of anterior instrumentation for scoliotic deformity. Case report. Neurosurg Focus 2012; 7:e2. [PMID: 16918210 DOI: 10.3171/foc.1999.7.5.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Correction of rigid scoliotic deformities involving the thoracic spine has required that a thoracotomy be performed to obtain anterior release to mobilize the deformity, as well as placement of corrective spinal instrumentation either via a separate posterior or anterior thoracic approach. To the best of the author's knowledge, this is the first published report of a case in which anterior correction of a deformity was achieved endoscopically. A 27-year-old man presented with a rigid 85 degrees thoracic kyphoscoliotic deformity that had developed over several years. He had previously undergone a C7-T12 laminectomy to decompress the spinal cord from a lipoma. Using thoracoscopic techniques, the author performed an anterior release and interbody fusion. Endoscopically, an anterior screw/rod system applied from T-5 to T-9 corrected the deformity to 55 degrees . There were no surgery-related complications. At follow-up examiniation 1.5 years after surgery, the patient had developed a solid fusion and the correction was maintained at an angle of 58 degrees . It is feasible to use thoracoscopic techniques to perform an anterior release and to apply anterior corrective spinal instrumentation to treat thoracic scoliotic deformities, thereby avoiding the need for an open posterior approach in which instrumentation is placed.
Collapse
Affiliation(s)
- C A Dickman
- Division of Neurological Surgery, Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
16
|
Awad H, Elgharably H, Popovich PG. Role of induced hypothermia in thoracoabdominal aortic aneurysm surgery. Ther Hypothermia Temp Manag 2012; 2:119-37. [PMID: 24716449 DOI: 10.1089/ther.2012.0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
For more than 50 years, hypothermia has been used in aortic surgery as a tool for neuroprotection. Hypothermia has been introduced into thoracoabdominal aortic aneurysm (TAAA) surgery by many cardiovascular centers to protect the body's organs, including the spinal cord. Numerous publications have shown that hypothermia can prevent immediate and delayed motor dysfunction after aortic cross-clamping. Here, we reviewed the historical application of hypothermia in aortic surgery, role of hypothermia in preclinical studies, cellular and molecular mechanisms by which hypothermia confers neuroprotection, and the role of systemic and regional hypothermia in clinical protocols to reduce and/or eliminate the devastating consequences of ischemic spinal cord injury after TAAA repair.
Collapse
Affiliation(s)
- Hamdy Awad
- 1 Department of Anesthesiology, Wexner Medical Center at The Ohio State University , Columbus, Ohio
| | | | | |
Collapse
|
17
|
Ji YB, Wu YM, Ji Z, Song W, Xu SY, Wang Y, Pan SY. Interrupted intracarotid artery cold saline infusion as an alternative method for neuroprotection after ischemic stroke. Neurosurg Focus 2012; 33:E10. [DOI: 10.3171/2012.5.focus1215] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Intracarotid artery cold saline infusion (ICSI) is an effective method for protecting brain tissue, but its use is limited because of undesirable secondary effects, such as severe decreases in hematocrit levels, as well as its relatively brief duration. In this study, the authors describe and investigate the effects of a novel ICSI pattern (interrupted ICSI) relative to the traditional method (uninterrupted ICSI).
Methods
Ischemic strokes were induced in 85 male Sprague-Dawley rats by occluding the middle cerebral artery for 3 hours using an intraluminal filament. Uninterrupted infusion groups received an infusion at 15 ml/hour for 30 minutes continuously. The same infusion speed was used in the interrupted infusion groups, but the whole duration was divided into trisections, and there was a 20-minute interval without infusion between sections. Forty-eight hours after reperfusion, H & E and silver nitrate staining were utilized for morphological assessment. Infarct sizes and brain water contents were determined using H & E staining and the dry-wet weight method, respectively. Levels of neuron-specific enolase (NSE), S100β protein, and matrix metalloproteinase 9 (MMP-9) in the serum were determined using enzyme-linked immunosorbent assay. Neurological deficits were also evaluated.
Results
Histology showed that interrupted ICSI did not affect neurons or fibers in rat brains, which suggests that this method is safe for brain tissues with ischemia. The duration of hypothermia induced by interrupted ICSI was longer than that induced via the traditional method, and the decrease in hematocrit levels was less pronounced. There were no differences in infarct size or brain water content between uninterrupted and interrupted ICSI groups, but neuron-specific enolase and matrix metalloproteinase 9 serum levels were more reduced after interrupted ICSI than after the traditional method.
Conclusions
Interrupted ICSI is a safe method. Compared with traditional ICSI, the interrupted method has a longer duration of hypothermia and less effect on hematocrit and offers more potentially improved neuroprotection, thereby making it more attractive as an infusion technique in the clinic.
Collapse
|
18
|
Yue X, Qiao D, Wang A, Tan X, Li Y, Liu C, Wang H. CD200 attenuates methamphetamine-induced microglial activation and dopamine depletion. ACTA ACUST UNITED AC 2012; 32:415-421. [PMID: 22684568 DOI: 10.1007/s11596-012-0072-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Indexed: 12/21/2022]
Abstract
This study examined the neuroprotective effect of cluster of differentiation molecule 200 (CD200) against methamphetamine (METH)-induced neurotoxicity. In the in vitro experiment, neuron-microglia cultures were treated with METH (20 μmol/L), METH (20 μmol/L)+CD200-Fc (10 μg/mL) or CD200-Fc (10 μg/mL). Those untreated served as control. Microglia activation expressed as the ratio of MHC-II/CD11b was assessed by flow cytometry. The cytokines (IL-1β, TNF-α) secreted by activated microglia were detected by enzyme-linked immunosorbent assay (ELISA). In the in vivo experiment, 40 SD rats were divided into control, METH, METH+CD200-Fc and CD200-Fc groups at random. Rats were intraperitoneally injected with METH (15 mg/kg 8 times at 12 h interval) in METH group, with METH (administered as the same dose and time as the METH group) and CD200-Fc (1 mg/kg at day 0, 2, 4 after METH injection) in METH+CD200-Fc group, with CD200-Fc (1 mg/kg injected as the same time as the METH+CD200-Fc group) or with physiological saline solution in the control group. The level of striatal dopamine (DA) in rats was measured by high-performance liquid chromatography (HPLC). The microglial cells were immunohistochemically detected for the expression of Iba-1, a marker for microglial activation. The results showed that METH could increase the microglia activation in the neuron-microglia cultures and elevate the secretion of IL-1β and TNF-α, which could be attenuated by CD200-Fc. Moreover, CD200-Fc could partially reverse the striatal DA depletion induced by METH and reduce the number of activated microglia, i.e. Iba-1-positive cells. It was concluded that CD200 may have neuroprotective effects against METH-induced neurotoxicity by inhibiting microglial activation and reversing DA depletion in striatum.
Collapse
Affiliation(s)
- Xia Yue
- Department of Forensic Science, Southern Medical University, Guangzhou, 510515, China
| | - Dongfang Qiao
- Department of Forensic Science, Southern Medical University, Guangzhou, 510515, China
| | - Aifeng Wang
- Department of Forensic Science, Southern Medical University, Guangzhou, 510515, China
| | - Xiaohui Tan
- Department of Forensic Science, Southern Medical University, Guangzhou, 510515, China
| | - Yanhong Li
- Department of Forensic science, Nanchang University, Nanchang, 330006, China
| | - Chao Liu
- Guangzhou Criminal Science & Technology Institute, Guangzhou, 510030, China
| | - Huijun Wang
- Department of Forensic Science, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
19
|
Body cooling ameliorating spinal cord injury may be neurogenesis-, anti-inflammation- and angiogenesis-associated in rats. ACTA ACUST UNITED AC 2011; 70:885-93. [PMID: 20693909 DOI: 10.1097/ta.0b013e3181e7456d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Body cooling (BC) or mild hypothermia therapy (about 33°C) is reportedly effective for spinal cord injury (SCI). However, the mechanisms underlying the beneficial effects of BC remain unclear, so does BC ameliorating SCI via promoting neurogenesis, anti-inflammation, and angiogenesis. METHODS The standard rat compression SCI model was tested hypothetically in two groups: one receiving BC (33°C) and the other, normothermia (37°C). Afterward, the effects of BC therapy on the hind limb locomotion, spinal cord infarction and apoptosis, angiogenesis, neurogenesis, and inflammation in these two groups of SCI were assessed. The other group of sham SCI was used as controls. RESULTS Apoptosis (evidenced by higher numbers of terminal deoxynucleotidyl- transferase-mediated and duDP-biotin nick end-labeling-positive cells), infarct, activated inflammation (evidenced by higher levels of tumor necrosis factor-α, interleukin-1β, and myeloperoxidase), and hind limb locomotor dysfunction were inspected in the untreated (37°C) SCI rats 4 days after SCI. When compared with those of untreated SCI rats, SCI rats receiving BC (33°C) displayed lower levels of apoptosis, infarct volume, activated inflammation, and hind limb locomotor dysfunction. In addition, that BC promoted both angiogenesis (evidenced by increased numbers of both vascular endothelial growth factors and bromodeoxyuridine-positive endothelial cells) and neurogenesis (evidenced by increased numbers of both glial cell line-derived neurotrophic growth factors and bromodeoxyuridine-neuronal-specific nuclear protein double positive cells) in the injured spinal cord was evaluated 4 days after SCI. CONCLUSION BC (33°C) improved SCI outcomes by promoting angiogenesis, neurogenesis, and anti-inflammation in a rat SCI model.
Collapse
|
20
|
Intra-arrest rapid head cooling improves postresuscitation myocardial function in comparison with delayed postresuscitation surface cooling. Crit Care Med 2010; 36:S434-9. [PMID: 20449907 DOI: 10.1097/ccm.0b013e31818a88b6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To compare resuscitation outcomes and myocardial function among intra-arrest head cooling, delayed surface cooling, and uncooled controls. DESIGN Prospective animal study. SETTING University-affiliated animal research laboratory. SUBJECTS Twenty-four male domestic pigs. INTERVENTIONS Ventricular fibrillation remained untreated for 10 mins after which animals were assigned into three groups: 1) intra-arrest head cooling, 2) postresuscitation surface cooling, and 3) uncooled controls. Head cooling by evaporative perfluorochemical began coincident with the start of cardiopulmonary resuscitation and continued for a total of 4 hrs. Surface cooling using a cooling blanket began at 2 hrs after return of spontaneous circulation and continued for 8 hrs. Control animals were treated identically with the exception for cooling. MEASUREMENTS AND MAIN RESULTS Return of spontaneous circulation was achieved in eight of eight head-cooled animals, in seven of eight surface-cooled animals, and in seven of eight of controls. Myocardial functions measured by transthoracic echocardiography were significantly better in the head-cooled animals than in surface-cooled and controls. All head-cooled animals survived for more than 96 hrs. This contrasted with six of eight survivors after surface cooling, and only two of eight among controls. CONCLUSIONS Both intra-arrest head cooling and delayed surface cooling improved postresuscitation myocardial dysfunction. The beneficial effects were greatest with head cooling initiated with cardiopulmonary resuscitation.
Collapse
|
21
|
|
22
|
Childs C. Human brain temperature: regulation, measurement and relationship with cerebral trauma: Part 1. Br J Neurosurg 2009; 22:486-96. [DOI: 10.1080/02688690802245541] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Xiong M, Yang Y, Chen GQ, Zhou WH. Post-ischemic hypothermia for 24h in P7 rats rescues hippocampal neuron: association with decreased astrocyte activation and inflammatory cytokine expression. Brain Res Bull 2009; 79:351-7. [PMID: 19406216 DOI: 10.1016/j.brainresbull.2009.03.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 03/27/2009] [Accepted: 03/27/2009] [Indexed: 12/17/2022]
Abstract
Hypothermia is an effective method for reducing the neuronal damage induced by hypoxia-ischemia (HI) but the underlying mechanism remains unclear. To investigate the effects of post-HI hypothermia on the developing brain, 7-day-old rats were subjected to left carotid artery ligation followed by 8% oxygen for 2h. They were divided into a hypothermia group (rectal temperature 32-33 degrees C for 24h) and a normothermia group (36-37 degrees C for 24h) immediately after hypoxia-ischemia. Animals were sacrificed at 12, 24 and 72 h for gene analysis and 0, 1, 3 and 7 days for protein analysis after HI. There was a significant decrease in infarct volume in the hypothermia group at 7 days after HI compared with that in the normothermia group. The hypothermia group had more neuronal nuclei (NeuN) positive neurons and lower levels of glial fibrillary acidic protein (GFAP) mRNA and immunoreactivity in the hippocampus CA1 region than the normothermia group. Real-time PCR showed no significant difference in glial cell line-derived neurotrophic factor (GDNF) mRNA expression in the hippocampus in the two groups at various time points after HI. However, GDNF protein level was significantly increased in the hypothermia group. On the other hand, mRNA and protein levels of the inflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interleukin-6 (IL-6) were dramatically decreased in the hypothermia compared with the normothermia group. The present findings highlight an apparent association between inhibition of hippocampal neuron loss by hypothermia and decreased astrocytosis and inflammatory cytokine release after hypoxia-ischemia in the developing brain.
Collapse
Affiliation(s)
- Man Xiong
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai 201102, China.
| | | | | | | |
Collapse
|
24
|
Morino T, Ogata T, Takeba J, Yamamoto H. Microglia inhibition is a target of mild hypothermic treatment after the spinal cord injury. Spinal Cord 2008; 46:425-31. [DOI: 10.1038/sj.sc.3102163] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
25
|
Hultström M, Jansson L, Bodin B, Källskog O. Moderate hypothermia induces a preferential increase in pancreatic islet blood flow in anesthetized rats. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1438-43. [PMID: 17626132 DOI: 10.1152/ajpregu.00259.2007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of the study was to characterize the effects of induced moderate hypothermia on splanchnic blood flow, with particular reference to that of the pancreas and the islets of Langerhans. We also investigated how interference with the autonomic nervous system at different levels influenced the blood perfusion during hypothermia. For this purpose, hypothermia (body temperature of 28°C) was induced by external cooling, whereas normothermic (37.5°C) anesthetized Sprague-Dawley rats were used as controls. Some rats were pretreated with either propranolol, yohimbine, atropine, hexamethonium, or a bilateral abdominal vagotomy. Our findings suggest that moderate hypothermia elicits complex, organ-specific circulatory changes, with increased perfusion noted in the pylorus, as well as the whole pancreas and the pancreatic islets. The pancreatic islets maintain their high blood perfusion through mechanisms involving both sympathetic and parasympathetic mediators, whereas the increased pyloric blood flow is mediated through parasympathetic mechanisms. Renal blood flow was decreased, and this can be prevented by ganglionic blockade and is also influenced by β-adrenoceptors.
Collapse
Affiliation(s)
- Michael Hultström
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
26
|
Fukui O, Kinugasa Y, Fukuda A, Fukuda H, Tskitishvili E, Hayashi S, Song M, Kanagawa T, Hosono T, Shimoya K, Murata Y. Post-ischemic hypothermia reduced IL-18 expression and suppressed microglial activation in the immature brain. Brain Res 2006; 1121:35-45. [PMID: 17010950 DOI: 10.1016/j.brainres.2006.08.121] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 08/21/2006] [Accepted: 08/27/2006] [Indexed: 11/15/2022]
Abstract
Inflammation is an important factor for hypoxia-ischemia (HI) brain injury. Interleukin (IL)-18 is a proinflammatory cytokine which may be a contributor to injury in the immature brain after HI. To investigate the effects of post-HI hypothermia on IL-18 in the developing brain, 7-day-old rats were subjected to left carotid artery ligation followed by 8% oxygen for 60 min and divided into a hypothermia group (rectal temperature 32 degrees C for 24 h) and a normothermia group (36 degrees C for 24 h). The IL-18 mRNA was analyzed with real-time RT-PCR, and the protein level was analyzed by Western blot, and the location and source of IL-18 were assessed by immunohistochemistry. The significant increase of the IL-18 mRNA was observed in the ipsilateral hemispheres of the normothermia group at 24 h and 72 h after HI compared with controls, but the level in the ipsilateral hemispheres of the hypothermia group was significantly reduced at both time points, compared with the normothermia group, respectively. The IL-18 protein level in the ipsilateral hemispheres of the normothermia group significantly increased at 72 h after HI compared with controls, however, the protein level of the hypothermia group was significantly decreased, compared with the normothermia group. IL-18-positive cells were observed throughout the entire cortex, corpus callosum (CC) and striatum in the ipsilateral hemispheres of normothermia group at 72 h after HI, however, little positive cells were observed in the hypothermia group. Double labeling immunostaining found that most of the IL-18-positive cells were colocalized with lectin, which is a marker of microglia. The number of ameboid microglia (AM) in the normothermia group was significantly increased in cortex and CC, compared with the number in controls, but there were very few ramified microglia (RM) in these areas. In contrast, the number of AM in the hypothermia group was significantly decreased in cortex and CC, compared with the number in the normothermia group, and there were no significant differences in the number of AM and RM between the hypothermia group and controls. In conclusion, we found that IL-18 mRNA and the protein level were attenuated by post-HI hypothermia and that post-HI hypothermia may decrease microglia activation in the developing brain.
Collapse
Affiliation(s)
- On Fukui
- Department of Obstetrics and Gynecology, Osaka University School of Medicine, 2-2 Yamada-oka, Suita, 565-0871, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The rare but potentially devastating clinical syndrome of fulminant hepatic failure has as its components severe encephalopathy and finally cerebral edema, hemodynamic instability, renal failure, coagulopathy, profound metabolic disturbances and a particular susceptibility to bacterial and fungal infection. Despite advances in medical management, fulminant hepatic failure in its most severe form carries a high mortality rate unless urgent orthotopic liver transplantation is carried out. However, availability of cadaveric donor organs is limited and, due to the rapidly progressive clinical course in many cases, a substantial proportion of patients will die or develop contraindications to transplantation before the procedure can be performed. Consequently, recent interest has centred on living donor transplantation and the possibility of providing temporary liver support, either through auxiliary partial organ transplantation, extracorporeal perfusion or transplantation of hepatocytes, to allow time for either a liver graft to become available or native liver regeneration, on which spontaneous survival ultimately depends, to occur.
Collapse
Affiliation(s)
- Jelica Kurtovic
- Institute of Hepatology, Royal Free and University College Medical School, 69-75 Chenies Mews, London, WC1E 6HX, England
| | | | | |
Collapse
|
28
|
Sakurai T, Itoh K, Liu Y, Higashitsuji H, Sumitomo Y, Sakamaki K, Fujita J. Low temperature protects mammalian cells from apoptosis initiated by various stimuli in vitro. Exp Cell Res 2005; 309:264-72. [PMID: 16018998 DOI: 10.1016/j.yexcr.2005.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Revised: 06/10/2005] [Accepted: 06/10/2005] [Indexed: 10/25/2022]
Abstract
Mild hypothermia shows protective effects on patients with brain damage and cardiac arrest. To elucidate the molecular mechanisms underlying these effects, we examined the effects of low temperature (32 degrees C) on cells exposed to a variety of stress in vitro. We found that 32 degrees C suppressed induction of apoptosis by cytotoxic stimuli such as adriamycin, etoposide, thapsigargin, NaCl, H(2)O(2), and anti-Fas antibody. In adriamycin-treated BALB/3T3 cells, the down-shift in temperature from 37 degrees C to 32 degrees C increased the Bcl-xL protein level and decreased the mRNA level of Puma and mitochondrial translocation of Bax, suppressing caspase-9-mediated apoptosis. Furthermore, the protein level and stability of p53 were decreased, and its nuclear export was increased concomitant with Mdm2 mRNA upregulation. The low temperature effect was not observed in p53(-/-)/Mdm2(-/-) mouse embryonic fibroblasts, suggesting that the effect is mediated by suppression of the p53 pathway. In contrast, while thapsigargin-induced apoptosis was suppressed by the low temperature, no effect on the p53 protein level was observed. Furthermore, the survival rate of p53(-/-)/Mdm2(-/-) cells exposed to thapsigargin was increased when cultured at 32 degrees C compared with 37 degrees C. In conclusion, mild hypothermia protects cells from a variety of stress by p53-dependent and p53-independent mechanisms.
Collapse
Affiliation(s)
- Toshiharu Sakurai
- Department of Clinical Molecular Biology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
29
|
Tattersall GJ, Gerlach RM. Hypoxia progressively lowers thermal gaping thresholds in bearded dragons,Pogona vitticeps. J Exp Biol 2005; 208:3321-30. [PMID: 16109893 DOI: 10.1242/jeb.01773] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
SUMMARYMost animals, including reptiles, lower body temperature(Tb) under hypoxic conditions. Numerous physiological and behavioural traits significant to the regulation of Tb are altered by hypoxia in ways that suggest an orchestrated adjustment of Tb at a new and lower regulated level. We examined this matter in bearded dragons, Pogona vitticeps, a species of reptile that naturally exhibits open mouth gaping at high temperatures, presumably in order to promote evaporation and thus prevent or avoid further increases in Tb. The threshold for the onset of gaping (assessed as the temperature at which lizards spent 50% of their time gaping) was reduced from 36.9°C in normoxia to 35.5°C at 10% and 34.3°C at 6%O2. The overall magnitude or degree of gaping, measured qualitatively, was more pronounced at lower temperatures in hypoxia. Females consistently had lower gaping threshold temperatures than did males, and this difference was retained throughout exposure to hypoxia. In addition to gaping,evaporative water loss from the cloaca may also play a significant role in temperature regulation, since the ambient temperature at which cloacal discharge occurred was also reduced significantly in hypoxia. The results reported herein strongly support the view that hypoxia reduces temperature set-point in lizards and that such changes are coordinated by specific behavioural thermoeffectors that modulate evaporative water loss and thus facilitate a high potential for controlling or modifying Tb.
Collapse
Affiliation(s)
- Glenn J Tattersall
- Department of Biological Sciences, Brock University, St Catharines, Ontario, L2S 3A1, Canada.
| | | |
Collapse
|
30
|
Zhao P, Zuo Z. Prenatal hypoxia-induced adaptation and neuroprotection that is inducible nitric oxide synthase-dependent. Neurobiol Dis 2005; 20:871-80. [PMID: 15994093 DOI: 10.1016/j.nbd.2005.05.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2004] [Revised: 03/20/2005] [Accepted: 05/20/2005] [Indexed: 10/25/2022] Open
Abstract
The incidence of perinatal stroke is approximately 0.025%. About two thirds of these patients develop long-lasting neurological deficits. Preconditioning-induced neuroprotection, a phenomenon in which application of a stimulus induces brain ischemic tolerance, is investigated to improve outcome after a perinatal stroke. We applied prenatal hypoxia to fetuses by exposing 22-day pregnant mother rats to 15% oxygen for 30 min and subjected newborns with or without this prenatal hypoxia to brain ischemia 48 h later. Newborns with the prenatal hypoxia had a lower mortality rate, less brain tissue and neuronal loss and fewer active caspase 3 (an indicator for cell apoptosis) positive brain cells than newborns with the brain ischemia only. This neuroprotection was abolished by an inhibitor of inducible nitric oxide synthase (iNOS). The expression of iNOS proteins but not endothelial and neuronal NOS proteins was increased by the prenatal hypoxia. Thus, the prenatal hypoxia-induced neuroprotection may be iNOS-dependent.
Collapse
Affiliation(s)
- Ping Zhao
- Department of Anesthesiology, Neuroscience and Neurological Surgery, University of Virginia, One Hospital Drive, PO Box 800710, Charlottesville, VA 22908-0710, USA
| | | |
Collapse
|
31
|
Thomas DM, Kuhn DM. MK-801 and dextromethorphan block microglial activation and protect against methamphetamine-induced neurotoxicity. Brain Res 2005; 1050:190-8. [PMID: 15987631 DOI: 10.1016/j.brainres.2005.05.049] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Revised: 05/19/2005] [Accepted: 05/22/2005] [Indexed: 11/22/2022]
Abstract
Methamphetamine causes long-term toxicity to dopamine nerve endings of the striatum. Evidence is emerging that microglia can contribute to the neuronal damage associated with disease, injury, or inflammation, but their role in methamphetamine-induced neurotoxicity has received relatively little attention. Lipopolysaccharide (LPS) and the neurotoxic HIV Tat protein, which cause dopamine neuronal toxicity after direct infusion into brain, cause activation of cultured mouse microglial cells as evidenced by increased expression of intracellular cyclooxygenase-2 and elevated secretion of tumor necrosis factor-alpha. MK-801, a non-competitive NMDA receptor antagonist that is known to protect against methamphetamine neurotoxicity, prevents microglial activation by LPS and HIV Tat. Dextromethorphan, an antitussive agent with NMDA receptor blocking properties, also prevents microglial activation. In vivo, MK-801 and dextromethorphan reduce methamphetamine-induced activation of microglia in striatum and they protect dopamine nerve endings against drug-induced nerve terminal damage. The present results indicate that the ability of MK-801 and dextromethorphan to protect against methamphetamine neurotoxicity is related to their common property as blockers of microglial activation.
Collapse
Affiliation(s)
- David M Thomas
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 2125 Scott Hall, 540 E. Canfield, Detroit, MI 48201, USA
| | | |
Collapse
|
32
|
Boddicker KA, Zhang Y, Zimmerman MB, Davies LR, Kerber RE. Hypothermia Improves Defibrillation Success and Resuscitation Outcomes From Ventricular Fibrillation. Circulation 2005; 111:3195-201. [PMID: 15956132 DOI: 10.1161/circulationaha.104.492108] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Induced hypothermia is recommended to improve neurological outcomes in unconscious survivors of out-of-hospital ventricular fibrillation (VF) cardiac arrest. Patients resuscitated from a VF arrest are at risk of refibrillation, but there are few data on the effects of already existing hypothermia on defibrillation and resuscitation.
Methods and Results—
Thirty-two swine (mean±SE weight, 23.0±0.6 kg) were divided into 4 groups: normothermia (n=8), mild hypothermia (35°C) (n=8), moderate hypothermia (33°C) (n=8), and severe hypothermia (30°C) (n=8). Hypothermia was induced by surrounding the animal with ice, and VF was electrically induced. After 8 minutes of unsupported VF (no CPR), the swine were defibrillated (biphasic waveform) with successive shocks as needed and underwent CPR until resumption of spontaneous circulation or no response (≥10 minutes). First-shock defibrillation success was higher in the moderate hypothermia group (6 of 8 hypothermia versus 1 of 8 normothermia;
P
=0.04). The number of shocks needed for late defibrillation (≥1 minute after initial shock) was less in all 3 hypothermia groups compared with normothermia (all
P
<0.05). None of the 8 animals in the normothermia group achieved resumption of spontaneous circulation compared with 3 of 8 mild hypothermia (
P
=NS), 7 of 8 moderate hypothermia (
P
=0.001), and 5 of 8 severe hypothermia (
P
=0.03) animals. Coronary perfusion pressure during CPR was not different between the groups.
Conclusions—
When VF was induced in the setting of moderate or severe hypothermia, resuscitative measures were facilitated with significantly improved defibrillation success and resuscitation outcome. The beneficial effect of hypothermia was not due to alteration of coronary perfusion pressure, which suggests that changes in the mechanical, metabolic, or electrophysiological properties of the myocardium may be responsible.
Collapse
|
33
|
Feiner JR, Bickler PE, Estrada S, Donohoe PH, Fahlman CS, Schuyler JA. Mild hypothermia, but not propofol, is neuroprotective in organotypic hippocampal cultures. Anesth Analg 2005; 100:215-225. [PMID: 15616081 DOI: 10.1213/01.ane.0000142129.17005.73] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The neuroprotective potency of anesthetics such as propofol compared to mild hypothermia remains undefined. Therefore, we determined whether propofol at two clinically relevant concentrations is as effective as mild hypothermia in preventing delayed neuron death in hippocampal slice cultures (HSC). Survival of neurons was assessed 2 and 3 days after 1 h oxygen and glucose deprivation (OGD) either at 37 degrees C (with or without 10 or 100 microM propofol) or at an average temperature of 35 degrees C during OGD (mild hypothermia). Cell death in CA1, CA3, and dentate neurons in each slice was measured with propidium iodide fluorescence. Mild hypothermia eliminated death in CA1, CA3, and dentate neurons but propofol protected dentate neurons only at a concentration of 10 microM; the more ischemia vulnerable CA1 and CA3 neurons were not protected by either 10 microM or 100 microM propofol. In slice cultures, the toxicity of 100 muM N-methyl-D-aspartate (NMDA), 500 microM glutamate, and 20 microM alpha-amino-5-methyl-4-isoxazole propionic acid (AMPA) was not reduced by 100 microM propofol. Because propofol neuroprotection may involve gamma-aminobutyric acid (GABA)-mediated indirect inhibition of glutamate receptors (GluRs), the effects of propofol on GluR activity (calcium influx induced by GluR agonists) were studied in CA1 neurons in HSC, in isolated CA1 neurons, and in cortical brain slices. Propofol (100 and 200 microM, approximate burst suppression concentrations) decreased glutamate-mediated [Ca2+]i increases (Delta[Ca2+]i) responses by 25%-35% in isolated CA1 neurons and reduced glutamate and NMDA Delta[Ca2+]i in acute and cultured hippocampal slices by 35%-50%. In both CA1 neurons and cortical slices, blocking GABAA receptors with picrotoxin reduced the inhibition of GluRs substantially. We conclude that mild hypothermia, but not propofol, protects CA1 and CA3 neurons in hippocampal slice cultures subjected to oxygen and glucose deprivation. Propofol was not neuroprotective at concentrations that reduce glutamate and NMDA receptor responses in cortical and hippocampal neurons.
Collapse
Affiliation(s)
- John R Feiner
- Department of Anesthesia and Perioperative Care, University of California, San Francisco
| | | | | | | | | | | |
Collapse
|
34
|
Hachimi-Idrissi S, Van Hemelrijck A, Michotte A, Smolders I, Sarre S, Ebinger G, Huyghens L, Michotte Y. Postischemic mild hypothermia reduces neurotransmitter release and astroglial cell proliferation during reperfusion after asphyxial cardiac arrest in rats. Brain Res 2004; 1019:217-25. [PMID: 15306256 DOI: 10.1016/j.brainres.2004.06.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2004] [Indexed: 12/15/2022]
Abstract
The present study investigated whether postischemic mild hypothermia attenuates the ischemia-induced striatal glutamate (GLU) and dopamine (DA) release, as well as astroglial cell proliferation in the brain. Anesthetized rats were exposed to 8 min of asphyxiation, including 5 min of cardiac arrest. The cardiac arrest was reversed to restoration of spontaneous circulation (ROSC), by brief external heart massage and ventilation within a period of 2 min. After the insult and during reperfusion, the extracellular glutamate and dopamine overflow increased to, respectively, 3000% and 5000% compared with the baseline values in the normothermic group and resulted in brain damage, ischemic neurons and gliosis. However, when hypothermia was induced for a period of 60 min after the insult and restoration of spontaneous circulation, the glutamate and dopamine overflows were not significantly different from that in the sham group. Histological analysis of the brain showed that postischemic mild hypothermia reduced brain damage, ischemic neurons, as well as astroglial cell proliferation. Thus, postischemic mild hypothermia reduces the excitotoxic process, brain damage, as well as astroglial cell proliferation during reperfusion. Moreover, these results emphasize the trigger effect of dopamine on the excitotoxic pathway.
Collapse
Affiliation(s)
- S Hachimi-Idrissi
- Department of Critical Care Medicine and Cerebral Resuscitation Research Group, van de Vrije Universiteit Brussel, Laarbeeklaan 101, Brussels B-1090, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ding Y, Li J, Luan X, Lai Q, McAllister JP, Phillis JW, Clark JC, Guthikonda M, Diaz FG. Local saline infusion into ischemic territory induces regional brain cooling and neuroprotection in rats with transient middle cerebral artery occlusion. Neurosurgery 2004; 54:956-64; discussion 964-5. [PMID: 15046664 DOI: 10.1227/01.neu.0000114513.96704.29] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2003] [Accepted: 11/18/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The neuroprotective effect of hypothermia has long been recognized. Use of hypothermia for stroke therapy, which is currently being induced by whole-body surface cooling, has been limited primarily because of management problems and severe side effects (e.g., pneumonia). The goal of this study was to determine whether local infusion of saline into ischemic territory could induce regional brain cooling and neuroprotection. METHODS A novel procedure was used to block the middle cerebral artery of rats for 3 hours with a hollow filament and locally infuse the middle cerebral artery-supplied territory with 6 ml cold saline (20 degrees C) for 10 minutes before reperfusion. RESULTS The cold saline infusion rapidly and significantly reduced temperature in cerebral cortex from 37.2 +/- 0.1 to 33.4 +/- 0.4 degrees C and in striatum from 37.5 +/- 0.2 to 33.9 +/- 0.4 degrees C. The significant hypothermia remained for up to 60 minutes after reperfusion. Significant (P < 0.01) reductions in infarct volume (approximately 90%) were evident after 48 hours of reperfusion. In ischemic rats that received the same amount of cold saline systemically through a femoral artery, a mild hypothermia was induced only in the cerebral cortex (35.3 +/- 0.2 degrees C) and returned to normal within 5 minutes. No significant reductions in infarct volume were observed in this group or in the ischemic group with local warm saline infusion or without infusion. Furthermore, brain-cooling infusion significantly (P < 0.01) improved motor behavior in ischemic rats after 14 days of reperfusion. This improvement continued for up to 28 days after reperfusion. CONCLUSION Local prereperfusion infusion effectively induced hypothermia and ameliorated brain injury from stroke. Clinically, this procedure could be used in acute stroke treatment, possibly in combination with intra-arterial thrombolysis or mechanical disruption of clot by means of a microcatheter.
Collapse
Affiliation(s)
- Yuchuan Ding
- Department of Neurological Surgery, Wayne State University School of Medicine, Lande Medical Research Building, Room 48, 550 East Canfield, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Hypothermia has been shown to be cerebroprotective in traumatized brains. Although a large number of traumatic brain injury (TBI) studies in animals have shown that hypothermia is effective in suppressing a variety of damaging mechanisms, clinical investigations have shown less consistent results. The complexity of damaging mechanisms in human TBI may contribute to these discrepancies. In particular, secondary injuries such as hypotension and hypoxemia may promote poor outcome. However, few experimental TBI studies have employed complex models that included such secondary injuries to clarify the efficacy of hypothermia. This review discusses the effects of hypothermia in various TBI models addressing primary and acute secondary injuries. Included are recently published clinical data using hypothermia as a therapeutic tool for preventing or reducing the detrimental posttraumatic secondary injuries and neurobehavioral deficits. Also discussed are recent successful applications of hypothermia from outside the TBI realm. Based on all available data, some general considerations for the application of hypothermia in TBI patients are given.
Collapse
Affiliation(s)
- Harald G Fritz
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Jena, Germany.
| | | |
Collapse
|
37
|
Luan X, Li J, McAllister JP, Diaz FG, Clark JC, Fessler RD, Ding Y. Regional brain cooling induced by vascular saline infusion into ischemic territory reduces brain inflammation in stroke. Acta Neuropathol 2004; 107:227-34. [PMID: 14691633 DOI: 10.1007/s00401-003-0802-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2003] [Revised: 11/13/2003] [Accepted: 11/13/2003] [Indexed: 12/22/2022]
Abstract
The neuroprotective effect of hypothermia has long been recognized. Use of hypothermia for stroke therapy, which is currently being induced by whole body surface cooling, has been largely limited because of management problems and severe side effects (i.e., pneumonia). Our recent studies have demonstrated the significant therapeutic value of local brain cooling in the ischemic territory prior to reperfusion in stroke. The goal of this study was to determine if cerebral local cooling infusion could reduce stroke-mediated brain injury by inhibiting inflammatory responses. A hollow filament was used to block the middle cerebral artery (MCA) for 3 hours, and then to locally infuse the ischemic territory with 6 ml cold saline (20 degrees C) for 10 min prior to 48-h reperfusion. This cold saline infusion significantly ( P<0.01) reduced temperature of the MCA supplied territory (in cerebral cortex from 37.2+/-0.1 degrees C to 33.4+/-0.4 degrees C, in striatum from 37.5+/-0.2 degrees C to 33.9+/-0.4 degrees C), with the hypothermia remaining for at least 45 min after reperfusion. Consequently, significant ( P<0.01) reductions in endothelial expression of intracellular adhesion molecule-1 (ICAM-1), the key step for inflammatory progress, as well as leukocyte infiltration, were evident in both cortex and striatum after reperfusion. As a control, ischemic rats received the same amount of cold saline systemically through a femoral artery. A mild hypothermia was induced in the cerebral cortex (35.3+/-0.2 degrees C) but not in the striatum (36.8+/-0.2 degrees C). The reduced cortical temperature returned to normal within 5 min. Brain temperature in ischemic rats perfused locally with saline at 37 degrees C remained normal. Intensive expression of ICAM-1 and accumulation of leukocytes was observed in ischemic control groups without brain cooling infusion. In conclusion, brain hypothermia induced by local pre-reperfusion infusion ameliorated brain inflammation from stroke.
Collapse
Affiliation(s)
- Xiaodong Luan
- Department of Neurological Surgery, Wayne State University School of Medicine, Lande Medical Research Building, Room 48, 550 E. Canfield, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Yosunkaya A, Ak A, Barişkaner H, Ustün ME, Tuncer S, Gürbilek M. Effect of Gamma-Hydroxybutyric Acid on Lipid Peroxidation and Tissue Lactate Level in Experimental Head Trauma. ACTA ACUST UNITED AC 2004; 56:585-90. [PMID: 15128130 DOI: 10.1097/01.ta.0000058119.60074.25] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND This study was designed to determine the effects of gamma-hydroxybutyric acid (GHB) on tissue lactate and malondialdehyde (MDA) levels in rabbit brain after experimental head trauma. METHODS Thirty New Zealand rabbits were divided equally into three groups: group S was the sham-operated group, group C, and group GHB received head trauma, where group C was the untreated and group GHB was the treated group. Head trauma was delivered by performing a craniectomy over the right hemisphere and dropping a weight of 10 g from a height of 80 cm. GHB was administered 400 mg/kg intravenously for 10 minutes after the head trauma to group GHB. The nontraumatized side was named "1" and the traumatized side was named "2." One hour after trauma, brain cortices were resected from both sides and the concentrations of lactate and MDA were determined. RESULTS There were significant differences between lactate and MDA levels of group S and all other groups (C1, C2, GHB1, and GHB2) except between lactate levels of group S and group GHB1, the nontraumatized and traumatized sides of groups C and group GHB, group C2 versus group GHB2, and group C1 versus group GHB1 (p < 0.05). Rectal temperature after the administration of GHB in group GHB was found lower than in groups S and C (p < 0.05). CONCLUSION These results demonstrate that head trauma leads to an increase in brain tissue lactate and MDA levels, and GHB effectively suppresses the increase of lactate and MDA.
Collapse
Affiliation(s)
- Alper Yosunkaya
- Department of Anesthesiology, Faculty of Meram Medicine, University of Selçuk, Konya, Turkey.
| | | | | | | | | | | |
Collapse
|
39
|
Chang CP, Lee CC, Chen SH, Lin MT. Aminoguanidine Protects Against Intracranial Hypertension and Cerebral Ischemic Injury in Experimental Heatstroke. J Pharmacol Sci 2004; 95:56-64. [PMID: 15153651 DOI: 10.1254/jphs.95.56] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The aim of the present study was to ascertain whether aminoguanidine attenuated intracranial hypertension and cerebral ischemic injury in experimental heatstroke. Urethane-anesthetized rats were exposed to heat stress (ambient temperature of 43 degrees C) to induce heatstroke. Control rats were exposed to 24 degrees C. Mean arterial pressure, cerebral perfusion pressure, and cerebral blood flow after the onset of heatstroke were all significantly lower than in control rats. However, colonic temperature, intracranial pressure, heart rate, cerebral inducible nitric oxide synthase (iNOS)-dependent NO, and neuronal damage score were greater after the onset of heatstroke. Aminoguanidine (30 micromol/kg, i.v.; 30 min before the start of heat exposure) pretreatment significantly attenuated the heatstroke-induced hyperthermia, arterial hypotension, intracranial hypertension, cerebral ischemia and neuronal damage, and increased iNOS-dependent NO formation in the brain. The extracellular concentrations of ischemic (e.g., glutamate and lactate/pyruvate ratio) and damage (e.g., glycerol) markers in the hypothalamus were also increased after the onset of heatstroke. Aminoguanidine pretreatment significantly attenuated the increase in hypothalamic ischemia and damage markers associated with heatstroke. Delaying onset of aminoguanidine administration (i.e., 0 or 30 min after the start of heat exposure) reduced the preventive efficiency on heatstroke-induced hyperthermia, arterial hypotension, intracranial hypertension, cerebral ischemia, and increased iNOS-dependent NO formation in brain. These results suggest that aminoguanidine protects against heatstroke-induced intracranial hypertension and cerebral ischemic injury by inhibition of cerebral iNOS-dependent NO production.
Collapse
Affiliation(s)
- Ching-Ping Chang
- Institute of Physiology, National Yang-Ming University Medical School, Taipei, Taiwan
| | | | | | | |
Collapse
|
40
|
Li L, Feng Z, Porter AG. JNK-dependent phosphorylation of c-Jun on serine 63 mediates nitric oxide-induced apoptosis of neuroblastoma cells. J Biol Chem 2003; 279:4058-65. [PMID: 14617628 DOI: 10.1074/jbc.m310415200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
c-Jun NH2-terminal kinases (JNKs) potentiate transcriptional activity of c-Jun by phosphorylating serines 63 and 73. Moreover, JNK and c-Jun can modulate apoptosis. However, an involvement of nitric oxide (NO)-induced phosphorylation of c-Jun on Ser-63 and Ser-73 in apoptosis has not been explored. We report that in SH-Sy5y neuroblastoma cells, NO induced apoptosis following JNK activation and phosphorylation of c-Jun almost exclusively on Ser-63. Importantly, NO-induced apoptosis and caspase-3 activity were inhibited in cells stably transformed with dominant-negative c-Jun in which Ser-63 is mutated to alanine (S63A), but not in cells transformed with dominant-negative c-Jun (S73A). Ser-63 of c-Jun (but not Ser-73) was required for NO-induced, c-Jun-dependent transcriptional activity. NO-induced apoptosis, Ser-63 phosphorylation of c-Jun, and caspase-3 activity were all inhibited in SH-Sy5y cells transformed with dominant-negative jnk. A caspase-3 inhibitor prevented apoptosis but not c-Jun phosphorylation. In a different neuroblastoma cell line, NO-induced Ser-63 phosphorylation of c-Jun and apoptosis were blocked by a specific JNK inhibitor. We conclude that NO-inducible apoptosis is mediated by JNK-dependent Ser-63 phosphorylation of c-Jun upstream of caspase-3 activation in neuroblastoma cells.
Collapse
Affiliation(s)
- Lei Li
- Institute of Molecular and Cell Biology, Singapore 117609, Republic of Singapore
| | | | | |
Collapse
|
41
|
Zheng S, Zuo Z. Isoflurane preconditioning reduces purkinje cell death in an in vitro model of rat cerebellar ischemia. Neuroscience 2003; 118:99-106. [PMID: 12676141 DOI: 10.1016/s0306-4522(02)00767-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We monitored survival of Purkinje cells in rat cerebellar slices to test the hypothesis that isoflurane preconditioning reduces ischemia-induced neuronal death. Preconditioning the brain slices with isoflurane, a volatile anesthetic commonly used in clinical practice, at 1-4% for 15 min at 37 degrees C significantly decreased Purkinje cell injury and death caused by a 20-min ischemia (simulated by oxygen-glucose deprivation, OGD). The effective concentration for half of the maximal effect (EC(50)) for this isoflurane preconditioning-induced neuroprotection was 1.17+/-0.31% and the maximal protective effects were achieved at 3% or higher concentrations of isoflurane. In addition, preconditioning the cells with isoflurane for 15-30 min was needed for the preconditioning to be maximally protective. Although farnesyl protein transferase inhibitor III blocked the protective effects of OGD preconditioning (a 3-min OGD 15 min before the 20-min OGD), this inhibitor did not affect the neuroprotection induced by isoflurane preconditioning. While DL-threo-beta-hydroxyaspartic acid (THA), a specific glutamate transporter inhibitor, did not change basal OGD-induced cell death rate, THA blocked the neuroprotection induced by isoflurane preconditioning but not by OGD preconditioning. Glybenclamide, a K(ATP) channel inhibitor, did not block the neuroprotection induced by either isoflurane or OGD preconditioning. Our results suggest that isoflurane preconditioning is neuroprotective. The isoflurane concentrations and times needed for the preconditioning to be neuroprotective are clinically relevant. The mechanisms of this protection seem to involve modulation of glutamate transporter activity.
Collapse
Affiliation(s)
- S Zheng
- Department of Anesthesiology, University of Virginia Health System, One Hospital Drive, P.O. Box 800710, Charlottesville, VA 22908-0710, USA
| | | |
Collapse
|
42
|
Otawara Y, Ogasawara K, Kubo Y, Tomitsuka N, Ogawa A, Suzuki M. Brain and systemic temperature in patients with severe subarachnoid hemorrhage. SURGICAL NEUROLOGY 2003; 60:159-64; discussion 164. [PMID: 12900131 DOI: 10.1016/s0090-3019(03)00083-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The significance of brain temperature (BT) in patients with severe brain damage remains unclear. This study investigated the relationships between BT, systemic temperature (ST), and clinical outcome in patients with severe subarachnoid hemorrhage. METHODS Thirty-one comatose patients with severe subarachnoid hemorrhage underwent ventricular drainage immediately after admission. The ventricular catheter also allowed monitoring of BT. ST was continuously measured using a bladder catheter with thermistor probe. RESULTS BT at the start of the monitoring was lower than ST in four patients, and all died of brain swelling. BT was higher than ST at first but later fell below ST ("temperature reversal") in 12 patients, who all died of acute brain swelling. BT was higher than ST throughout the monitoring in 15 patients. Five of these patients died of causes other than brain swelling such as rerupture of the cerebral aneurysm, multiple organ failure, or respiratory failure. The other 10 patients survived with various degrees of disability. CONCLUSIONS Observation of BT and ST can predict the outcome of severe subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Yasunari Otawara
- Department of Neurosurgery, Iwate Medical University, Morioka, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Chou YT, Lai ST, Lee CC, Lin MT. Hypothermia attenuates circulatory shock and cerebral ischemia in experimental heatstroke. Shock 2003; 19:388-93. [PMID: 12688553 DOI: 10.1097/00024382-200304000-00016] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We tested the hypothesis in a rat model that body cooling suppresses circulatory shock and cerebral ischemia in heatstroke. Animals under urethane anesthesia were exposed to water blanket temperature (Tblanket) of 42 degrees C until mean arterial pressure (MAP) and local cerebral blood flow (CBF) in the hippocampus began to decrease from their peak levels, which was arbitrarily defined as the onset of heatstroke. Control rats were exposed to 26 degrees C. Extracellular concentrations of glutamate, glycerol, lactate, and lactate/pyruvate in the hippocampus were assessed by microdialysis methods. Cooling was accomplished by decreasing Tblanket from 42 degrees C to 16 degrees C. The values of MAP and CBF after the onset of heat stroke in heatstroke rats received no cooling were all significantly lower than those in control rats. However, the neuronal damage score and extracellular levels of ischemia and damage markers in the hippocampus were greater. Cooling immediately after the onset of heatstroke reduced the heatstroke-induced circulatory shock, cerebral ischemia, neuronal damage, and surge of tissue ischemia and damage markers in the hippocampus, and resulted in prolongation of survival time. Delaying the onset of cooling reduced the therapeutic efficiency. The results suggest that body cooling attenuates circulatory shock and cerebral ischemia insults in heatstroke.
Collapse
Affiliation(s)
- Yueh-Ting Chou
- Institute of Physiology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | |
Collapse
|
44
|
Gibbons H, Sato TA, Dragunow M. Hypothermia suppresses inducible nitric oxide synthase and stimulates cyclooxygenase-2 in lipopolysaccharide stimulated BV-2 cells. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 110:63-75. [PMID: 12573534 DOI: 10.1016/s0169-328x(02)00585-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypothermia is neuroprotective, possibly through suppression of microglial activation. We investigated the effects of hypothermia on lipopolysaccharide (LPS) stimulated BV-2 cells. At 37 degrees C, LPS elicited strong increases in inducible nitric oxide synthase (iNOS), nitric oxide (NO), cyclooxygenase-2 (COX-2), tumour necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6), accompanied by translocation of nuclear factor-kappaB (NF-kappaB) to the nucleus. Hypothermia (33 degrees C) caused complete suppression of iNOS and NO, a partial reduction of IL-6 but did not prevent TNF-alpha production or NF-kappaB translocation. In contrast, LPS induced cyclooxygenase-2 (COX-2) to higher levels under hypothermic conditions. These results show that hypothermia selectively suppresses iNOS in microglia.
Collapse
Affiliation(s)
- Hannah Gibbons
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
45
|
Schmutzhard E, Engelhardt K, Beer R, Brössner G, Pfausler B, Spiss H, Unterberger I, Kampfl A. Safety and efficacy of a novel intravascular cooling device to control body temperature in neurologic intensive care patients: a prospective pilot study. Crit Care Med 2002; 30:2481-8. [PMID: 12441758 DOI: 10.1097/00003246-200211000-00013] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine the safety and efficacy of a novel intravascular cooling device (Cool Line catheter with Cool Gard system) to control body temperature (temperature goal <37 degrees C) in neurologic intensive care patients. DESIGN A prospective, uncontrolled pilot study in 51 consecutive neurologic intensive care patients. SETTING A neurologic intensive care unit at a tertiary care university hospital. PARTICIPANTS Patients were 51 neurologic intensive care patients with an intracranial disease requiring a central venous catheter due to the primary (intracranial) disease. We excluded patients under the age of 19 yrs and those with active cardiac arrhythmia, full sepsis syndrome, bleeding diathesis and infection, or bleeding at the site of the intended catheter insertion. Male to female ratio was 31:20, and the median age was 55 yrs (range, 24-85 yrs). Forty-four of 51 patients (86.3%) had an initial Glasgow Coma Scale score of 3, three patients had a Glasgow Coma Scale score of 9, one patient presented with an initial Glasgow Coma Scale score of 11, two patients had an initial Glasgow Coma Scale score of 13, and one patient had an initial Glasgow Coma Scale score of 15. The mean initial tissue injury severity score was 45.1 and the median initial tissue injury severity score 45.0 (range, 19-70). INTERVENTIONS Patients were enrolled prospectively in a consecutive way. Within 12 hrs after admission, the intravascular cooling device (Cool Line catheter) was placed, the temperature probe was located within the bladder (by Foley catheter), and the Cool Gard cooling device was initiated. This Cool Gard system circulates temperature-controlled sterile saline through two small balloons mounted on the distal end of the Cool Line catheter. The patient's blood is gently cooled as it is passed over the balloons. The Cool Gard system has been set with a target temperature of 36.5 degrees C. The primary purpose and end point of this study was to evaluate the cooling capacity of this intravascular cooling device. Efficacy is expressed by the calculation formula of fever burden, which is defined as the fever time product ( degrees C hours) under the fever curve. MEASUREMENTS AND MAIN RESULTS The cooling device was in operation for a mean of 152.4 hrs. The ease of insertion was judged as easy in 42 of 51 patients; in a single patient, the catheter was malpositioned within the jugular vein, requiring early removal. The rate of infectious and noninfectious complications (nosocomial pneumonia, bacteremia, catheter-related ventriculitis, pulmonary embolism, etc.) was comparable to the rate usually observed in our neurologic intensive care patients with such severe intracranial diseases. The total fever burden within the entire study period of (on average) 152.4 hrs was 4.0 degrees C hrs/patient, being equivalent to 0.6 degrees C hrs/patient and day. Thirty of 51 patients showed an elevation of the body temperature (>37.9 degrees C) within 24 hrs after termination of the cooling study. One awake patient (subarachnoid hemorrhage, Glasgow Coma Scale score 15) experienced mild to moderate shivering throughout the entire period of 7 days. The mortality rate was 23.5%. CONCLUSION This novel intravascular cooling device (Cool Line catheter and Cool Gard cooling device) was highly efficacious in prophylactically controlling the body temperature of neurologic intensive care patients with very severe intracranial disease (median Glasgow Coma Scale score, 3-15). Morbidity and mortality rates were consistent with the ranges reported in the literature for such neurologic intensive patients.
Collapse
|
46
|
Kinoshita K, Chatzipanteli IK, Vitarbo E, Truettner JS, Alonso OF, Dietrich WD. Interleukin-1beta messenger ribonucleic acid and protein levels after fluid-percussion brain injury in rats: importance of injury severity and brain temperature. Neurosurgery 2002; 51:195-203; discussion 203. [PMID: 12182417 DOI: 10.1097/00006123-200207000-00027] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Posttraumatic temperature manipulations have been reported to significantly influence the inflammatory response to traumatic brain injury (TBI). The purpose of this study was to determine the temporal and regional profiles of messenger ribonucleic acid (mRNA) expression and protein levels for the proinflammatory cytokine interleukin-1beta (IL-1beta), after moderate or severe TBI. The effects of posttraumatic hypothermia (33 degrees C) or hyperthermia (39.5 degrees C) on these consequences of TBI were then determined. METHODS Male Sprague-Dawley rats underwent fluid-percussion brain injury. In the first phase of the study, rats were killed 15 minutes or 1, 3, or 24 hours after moderate TBI (1.8-2.2 atmospheres), for reverse transcription-polymerase chain reaction analysis. Other groups of rats were killed 1, 3, 24, or 72 hours after moderate or severe TBI (2.4-2.7 atmospheres), for protein analysis. In the second phase, rats underwent moderate fluid-percussion brain injury, followed immediately by 3 hours of posttraumatic normothermia (37 degrees C), hyperthermia (39.5 degrees C), or hypothermia (33 degrees C), and were then killed, for analyses of protein levels and mRNA expression. Brain samples, including cerebral cortex, hippocampus, thalamus, and cerebellum, were dissected and stored at -80 degrees C until analyzed. RESULTS The findings indicated that mRNA levels were increased (P < 0.05) as early as 1 hour after TBI and remained elevated up to 3 hours after moderate TBI. Although both moderate and severe TBI induced increased levels of IL-1beta (P < 0.05), increased protein levels were also noted in remote brain structures after severe TBI. Posttraumatic hypothermia attenuated IL-1beta protein levels, compared with normothermia (P < 0.05), although the levels remained elevated in comparison with sham values. In contrast, hyperthermia had no significant effect on IL-1beta levels, compared with normothermic values. Posttraumatic temperature manipulations had no significant effect on IL-1beta mRNA levels. CONCLUSION Injury severity determines the degree of IL-1beta protein level elevation after TBI. The effects of posttraumatic hypothermia on IL-1beta protein levels (an important mediator of neurodegeneration after TBI) may partly explain the established effects of posttraumatic temperature manipulations on inflammatory processes after TBI.
Collapse
Affiliation(s)
- Kosaku Kinoshita
- Department of Neurological Surgery, The Neurotrauma Research Center, University of Miami School of Medicine, Florida 33136, USA
| | | | | | | | | | | |
Collapse
|
47
|
Vejchapipat P, Proctor E, Ramsay A, Petros A, Gadian DG, Spitz L, Pierro A. Intestinal energy metabolism after ischemia-reperfusion: Effects of moderate hypothermia and perfluorocarbons. J Pediatr Surg 2002; 37:786-90. [PMID: 11987102 DOI: 10.1053/jpsu.2002.32288] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE This study investigated the roles of moderate hypothermia and extraluminal oxygenated perfluorcarbon (PFC) on intestinal metabolism after ischemia-reperfusion. METHODS A model of 30-minute intestinal ischemia followed by 60 minutes of reperfusion was used. The animals were maintained at either normothermia (36.5 to 37.5 degrees C) or moderate hypothermia (31 to 32 degrees C). Four groups of adult rats were studied (n = 8 per group): (A) sham at normothermia, (B) ischemia-reperfusion at normothermia, (C) ischemia-reperfusion at hypothermia and, (D) ischemia-reperfusion with extraluminal oxygenated PFC perfusion during ischemia at normothermia. Intestinal phosphocreatine, ATP and lactate levels were measured. Histologic changes in the intestine were evaluated. RESULTS Intestinal ischemia-reperfusion at normothermia caused a marked reduction in phosphocreatine and ATP with an increase in lactate. Moderate hypothermia exerted beneficial effects by attenuating the depletion of high-energy phosphates and the elevation of lactate. Extraluminal PFC perfusion during ischemia failed to produce a protective effect on high-energy phosphates, although it reduced lactate accumulation. Moderate hypothermia significantly decreased the degree of mucosal damage. CONCLUSIONS Whole-body moderate hypothermia protects the small intestine from reperfusion injury as measured both biochemically and histologically. Extraluminal oxygenated PFC administration during ischemia did not protect the intestine from reperfusion injury in this model.
Collapse
|
48
|
|
49
|
Harada K, Maekawa T, Tsuruta R, Kaneko T, Sadamitsu D, Yamashima T, Yoshida Ki KI. Hypothermia inhibits translocation of CaM kinase II and PKC-alpha, beta, gamma isoforms and fodrin proteolysis in rat brain synaptosome during ischemia-reperfusion. J Neurosci Res 2002; 67:664-9. [PMID: 11891778 DOI: 10.1002/jnr.10159] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
To clarify the involvement of intracellular signaling pathway and calpain in the brain injury and its protection by mild hypothermia, immunoblotting analyses were performed in the rat brain after global forebrain ischemia and reperfusion. After 30 min of ischemia followed by 60 min of reperfusion, Ca2+/calmodulin-dependent kinase II (CaM kinase II) and protein kinase C (PKC)-alpha, beta, gamma isoforms translocated to the synaptosomal fraction, while mild hypothermia (32 degrees C) inhibited the translocation. The hypothermia also inhibited fodrin proteolysis caused by ischemia-reperfusion, indicating the inhibition of calpain. These effects of hypothermia may explain the mechanism of the protection against brain ischemia-reperfusion injury through modulating synaptosomal function.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Emergency and Critical Care Medicine, Yamaguchi University School of Medicine, Yamaguchi, Japan
| | | | | | | | | | | | | |
Collapse
|
50
|
Burgess N, Isert P. Anaesthetic considerations for patients undergoing hypothermic cardiopulmonary bypass for complex neurovascular lesions: case presentation and review. Anaesth Intensive Care 2001; 29:406-16. [PMID: 11512653 DOI: 10.1177/0310057x0102900413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The anaesthetic management of a 38-year-old woman having excision of a meningioma involving the superior sagittal sinus is described. The procedure was performed using low flow moderate hypothermic cardiopulmonary bypass with central cannulation. Relevant literature is reviewed.
Collapse
Affiliation(s)
- N Burgess
- Department of Anaesthesia and Intensive Care, Prince of Wales Hospital, Sydney, New South Wales
| | | |
Collapse
|