1
|
Gruenbaum BF, Merchant KS, Zlotnik A, Boyko M. Gut Microbiome Modulation of Glutamate Dynamics: Implications for Brain Health and Neurotoxicity. Nutrients 2024; 16:4405. [PMID: 39771027 PMCID: PMC11677762 DOI: 10.3390/nu16244405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/07/2024] [Accepted: 12/20/2024] [Indexed: 01/05/2025] Open
Abstract
The gut-brain axis plays an integral role in maintaining overall health, with growing evidence suggesting its impact on the development of various neuropsychiatric disorders, including depression. This review explores the complex relationship between gut microbiota and glutamate (Glu) regulation, highlighting its effect on brain health, particularly in the context of depression following certain neurological insults. We discuss how microbial populations can either facilitate or limit Glu uptake, influencing its bioavailability and predisposing to neuroinflammation and neurotoxicity. Additionally, we examine the role of gut metabolites and their influence on the blood-brain barrier and neurotransmitter systems involved in mood regulation. The therapeutic potential of microbiome-targeted interventions, such as fecal microbiota transplantation, is also highlighted. While much research has explored the role of Glu in major depressive disorders and other neurological diseases, the contribution of gut microbiota in post-neurological depression remains underexplored. Future research should focus on explaining the mechanisms linking the gut microbiota to neuropsychiatric outcomes, particularly in conditions such as post-stroke depression, post-traumatic brain-injury depression, and epilepsy-associated depression. Systematic reviews and human clinical studies are needed to establish causal relationships and assess the efficacy of microbiome-targeted therapies in improving the neuropsychiatric sequalae after neurological insults.
Collapse
Affiliation(s)
- Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Kiran S. Merchant
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (A.Z.); (M.B.)
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (A.Z.); (M.B.)
| |
Collapse
|
2
|
Freedman Jr. RJ, Schock RB, Peacock WF. Therapeutic hypothermia is not dead, but hibernating! Clin Exp Emerg Med 2024; 11:238-242. [PMID: 39390630 PMCID: PMC11467454 DOI: 10.15441/ceem.24.291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Affiliation(s)
| | - Robert B. Schock
- Division of Research, Development, and Manufacturing, Life Recovery Systems, Kinnelon, NJ, USA
| | - W. Frank Peacock
- Henry J.N. Taub Department of Emergency Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Li Y, Wu C, Yang R, Tang J, Li Z, Yi X, Fan Z. Application and Development of Cell Membrane Functionalized Biomimetic Nanoparticles in the Treatment of Acute Ischemic Stroke. Int J Mol Sci 2024; 25:8539. [PMID: 39126107 PMCID: PMC11313357 DOI: 10.3390/ijms25158539] [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: 05/25/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Ischemic stroke is a serious neurological disease involving multiple complex physiological processes, including vascular obstruction, brain tissue ischemia, impaired energy metabolism, cell death, impaired ion pump function, and inflammatory response. In recent years, there has been significant interest in cell membrane-functionalized biomimetic nanoparticles as a novel therapeutic approach. This review comprehensively explores the mechanisms and importance of using these nanoparticles to treat acute ischemic stroke with a special emphasis on their potential for actively targeting therapies through cell membranes. We provide an overview of the pathophysiology of ischemic stroke and present advances in the study of biomimetic nanoparticles, emphasizing their potential for drug delivery and precision-targeted therapy. This paper focuses on bio-nanoparticles encapsulated in bionic cell membranes to target ischemic stroke treatment. It highlights the mechanism of action and research progress regarding different types of cell membrane-functionalized bi-onic nanoparticles such as erythrocytes, neutrophils, platelets, exosomes, macrophages, and neural stem cells in treating ischemic stroke while emphasizing their potential to improve brain tissue's ischemic state and attenuate neurological damage and dysfunction. Through an in-depth exploration of the potential benefits provided by cell membrane-functionalized biomimetic nanoparticles to improve brain tissue's ischemic state while reducing neurological injury and dysfunction, this study also provides comprehensive research on neural stem cells' potential along with that of cell membrane-functionalized biomimetic nanoparticles to ameliorate neurological injury and dysfunction. However, it is undeniable that there are still some challenges and limitations in terms of biocompatibility, safety, and practical applications for clinical translation.
Collapse
Affiliation(s)
- Ying Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Chuang Wu
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Rui Yang
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Jiannan Tang
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhanqing Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Xue Yi
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhongxiong Fan
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
4
|
Mezuki S, Matsuo R, Irie F, Shono Y, Kuwashiro T, Sugimori H, Wakisaka Y, Ago T, Kamouchi M, Kitazono T. Body temperature in the acute phase and clinical outcomes after acute ischemic stroke. PLoS One 2024; 19:e0296639. [PMID: 38206979 PMCID: PMC10783745 DOI: 10.1371/journal.pone.0296639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND This study aimed to examine whether post-stroke early body temperature is associated with neurological damage in the acute phase and functional outcomes at three months. METHODS We included 7,177 patients with acute ischemic stroke within 24 h of onset. Axillary temperature was measured daily in the morning for seven days. Mean body temperature was grouped into five quintiles (Q1: 35.1‒36.5°C, Q2: 36.5‒36.7°C, Q3: 36.7‒36.8°C, Q4: 36.8‒37.1°C, and Q5: 37.1‒39.1°C). Clinical outcomes included neurological improvement during hospitalization and poor functional outcome (modified Rankin scale score, 3-6) at three months. A logistic regression analysis was performed to evaluate the association between body temperature and clinical outcomes. RESULTS The patient's mean (SD) age was 70.6 (12.3) years, and 35.7% of patients were women. Mean body temperature was significantly associated with less neurological improvement from Q2 (odds ratios [95% confidence interval], 0.77 [0.65-0.99] vs. Q1) to Q5 (0.33 [0.28-0.40], P for trend <0.001) even after adjusting for potential confounders, including baseline neurological severity, C-reactive protein levels, and post-stroke acute infections. The multivariable-adjusted risk of poor functional outcome linearly increased from Q2 (1.36 [1.03-1.79]) to Q5 (6.44 [5.19-8.96], P for trend <0.001). These associations were maintained even in the analyses excluding patients with acute infectious diseases. Multivariable-adjusted risk of poor functional outcome was higher in patients with early body temperature elevation on days 1-3 and with longer duration with body temperature >37.0°C. CONCLUSIONS Post-stroke early high body temperature is independently associated with unfavorable outcomes following acute ischemic stroke.
Collapse
Affiliation(s)
- Satomi Mezuki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Ryu Matsuo
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumi Irie
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Shono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Takahiro Kuwashiro
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cerebrovascular Medicine and Neurology, Kyushu Medical Center, Fukuoka, Japan
| | - Hiroshi Sugimori
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cerebrovascular Medicine and Neurology, Kyushu Medical Center, Fukuoka, Japan
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Kamouchi
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
5
|
Li X, Helleringer R, Martucci LL, Dallérac G, Cancela JM, Galante M. Low Temperature Delays the Effects of Ischemia in Bergmann Glia and in Cerebellar Tissue Swelling. Biomedicines 2023; 11:biomedicines11051363. [PMID: 37239034 DOI: 10.3390/biomedicines11051363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/23/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Cerebral ischemia results in oxygen and glucose deprivation that most commonly occurs after a reduction or interruption in the blood supply to the brain. The consequences of cerebral ischemia are complex and involve the loss of metabolic ATP, excessive K+ and glutamate accumulation in the extracellular space, electrolyte imbalance, and brain edema formation. So far, several treatments have been proposed to alleviate ischemic damage, yet few are effective. Here, we focused on the neuroprotective role of lowering the temperature in ischemia mimicked by an episode of oxygen and glucose deprivation (OGD) in mouse cerebellar slices. Our results suggest that lowering the temperature of the extracellular 'milieu' delays both the increases in [K+]e and tissue swelling, two dreaded consequences of cerebellar ischemia. Moreover, radial glial cells (Bergmann glia) display morphological changes and membrane depolarizations that are markedly impeded by lowering the temperature. Overall, in this model of cerebellar ischemia, hypothermia reduces the deleterious homeostatic changes regulated by Bergmann glia.
Collapse
Affiliation(s)
- Xia Li
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Romain Helleringer
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Lora L Martucci
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Glenn Dallérac
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - José-Manuel Cancela
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Micaela Galante
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
6
|
Li P, Sun Z, Tian T, Yu D, Tian H, Gong P. Recent developments and controversies in therapeutic hypothermia after cardiopulmonary resuscitation. Am J Emerg Med 2023; 64:1-7. [PMID: 36435004 DOI: 10.1016/j.ajem.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/23/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022] Open
Abstract
Therapeutic hypothermia was recommended as the only neuroprotective treatment in comatose patients after return of spontaneous circulation (ROSC). With new evidence suggesting a similar neuroprotective effect of 36 °C and 33 °C, the term "therapeutic hypothermia" was substituted by "targeted temperature management" in 2011, which in turn was replaced by the term "temperature control" in 2022 because of new evidence of the similar effects of target normothermia and 33 °C. However, there is no clear consensus on the efficacy of therapeutic hypothermia. In this article, we provide an overview of the recent evidence from basic and clinical research related to therapeutic hypothermia and re-evaluate its application as a post-ROSC neuroprotective intervention in clinical settings.
Collapse
Affiliation(s)
- Peijuan Li
- Department of Emergency, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Dalian Medical University, Dalian, Liaoning, China
| | - Zhangping Sun
- Department of Emergency, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Dalian Medical University, Dalian, Liaoning, China
| | - Tian Tian
- Department of Emergency, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Dalian Medical University, Dalian, Liaoning, China
| | - Dongping Yu
- Department of Emergency, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hui Tian
- Department of Emergency, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Ping Gong
- Department of Emergency, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China; Department of Emergency, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
7
|
The effects of hypothermia on glutamate and γ-aminobutyric acid metabolism during ischemia in monkeys: a repeated-measures ANOVA study. Sci Rep 2022; 12:14470. [PMID: 36008544 PMCID: PMC9411555 DOI: 10.1038/s41598-022-18783-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
During an ischemic stroke, the brain releases various factors, including glutamate and γ-aminobutyric acid. Glutamate can cause neurotoxic effects through certain receptors and exacerbate neurological damage, while γ-aminobutyric acid as an inhibitory neurotransmitter can antagonize the excitotoxic effects of glutamate and enhance the tolerance of neurons to ischemia. Therefore, in this study, the content of amino acid neurotransmitters in brain tissue before ischemia, after 10 min of ischemia, hypothermic perfusion, and rewarming were analyzed by high-performance liquid chromatography-UV in an animal model of ischemic stroke generated by blocking the bilateral common carotid arteries of rhesus monkeys. The changes in amino acid neurotransmitters in the rhesus monkey brain during post-ischemia hypothermic perfusion and rewarming were investigated by statistical methods of repeated measures ANOVA, showing that the concentration change of glutamate had not only a temporal factor but also was influenced by temperature, and there was an interaction effect between the two. Time but not temperature affected the change in γ-aminobutyric acid concentration, and there was an interaction effect between the two. Accordingly, hypoperfusion exerts a protective effect during ischemia by inhibiting the release of excitatory amino acid neurotransmitters, while the antagonistic effect of GABA on Glu is not significant.
Collapse
|
8
|
Abulseoud OA, Alasmari F, Hussein AM, Sari Y. Ceftriaxone as a Novel Therapeutic Agent for Hyperglutamatergic States: Bridging the Gap Between Preclinical Results and Clinical Translation. Front Neurosci 2022; 16:841036. [PMID: 35864981 PMCID: PMC9294323 DOI: 10.3389/fnins.2022.841036] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation of glutamate homeostasis is a well-established core feature of neuropsychiatric disorders. Extracellular glutamate concentration is regulated by glutamate transporter 1 (GLT-1). The discovery of a beta-lactam antibiotic, ceftriaxone (CEF), as a safe compound with unique ability to upregulate GLT-1 sparked the interest in testing its efficacy as a novel therapeutic agent in animal models of neuropsychiatric disorders with hyperglutamatergic states. Indeed, more than 100 preclinical studies have shown the efficacy of CEF in attenuating the behavioral manifestations of various hyperglutamatergic brain disorders such as ischemic stroke, amyotrophic lateral sclerosis (ALS), seizure, Huntington’s disease, and various aspects of drug use disorders. However, despite rich and promising preclinical data, only one large-scale clinical trial testing the efficacy of CEF in patients with ALS is reported. Unfortunately, in that study, there was no significant difference in survival between placebo- and CEF-treated patients. In this review, we discussed the translational potential of preclinical efficacy of CEF based on four different parameters: (1) initiation of CEF treatment in relation to induction of the hyperglutamatergic state, (2) onset of response in preclinical models in relation to onset of GLT-1 upregulation, (3) mechanisms of action of CEF on GLT-1 expression and function, and (4) non-GLT-1-mediated mechanisms for CEF. Our detailed review of the literature brings new insights into underlying molecular mechanisms correlating the preclinical efficacy of CEF. We concluded here that CEF may be clinically effective in selected cases in acute and transient hyperglutamatergic states such as early drug withdrawal conditions.
Collapse
Affiliation(s)
- Osama A. Abulseoud
- Department of Psychiatry and Psychology, Alex School of Medicine at Mayo Clinic, Phoenix, AZ, United States
- *Correspondence: Osama A. Abulseoud,
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Youssef Sari,
| |
Collapse
|
9
|
Usmanov ES, Chubarova MA, Saidov SK. Emerging Trends in the Use of Therapeutic Hypothermia as a Method for Neuroprotection in Brain Damage (Review). Sovrem Tekhnologii Med 2021; 12:94-104. [PMID: 34796010 PMCID: PMC8596265 DOI: 10.17691/stm2020.12.5.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Indexed: 11/14/2022] Open
Abstract
The review analyzes current clinical studies on the use of therapeutic hypothermia as a neuroprotective method for treatment of brain damage. This method yields good outcomes in patients with acute brain injuries and chronic critical conditions. There has been shown the interest of researchers in studying the preventive potential of therapeutic hypothermia in secondary neuronal damage. There has been described participation of new molecules producing positive effect on tissues and cells of the central nervous system - proteins and hormones of cold stress - in the mechanisms of neuroprotection in the brain. The prospects of using targeted temperature management in treatment of brain damage are considered.
Collapse
Affiliation(s)
- E Sh Usmanov
- Researcher, Laboratory of Clinical Neurophysiology; Federal Clinical Research Centre for Intensive Care Medicine and Rehabilitology, 777 Lytkino Village, Solnechnogorsk District, Moscow Region, 141534, Russia
| | - M A Chubarova
- Junior Researcher, Laboratory of Clinical Neurophysiology; Federal Clinical Research Centre for Intensive Care Medicine and Rehabilitology, 777 Lytkino Village, Solnechnogorsk District, Moscow Region, 141534, Russia
| | - Sh Kh Saidov
- Senior Researcher, Laboratory of Clinical Neurophysiology Federal Clinical Research Centre for Intensive Care Medicine and Rehabilitology, 777 Lytkino Village, Solnechnogorsk District, Moscow Region, 141534, Russia
| |
Collapse
|
10
|
Shevelev O, Petrova M, Smolensky A, Osmonov B, Toimatov S, Kharybina T, Karbainov S, Ovchinnikov L, Vesnin S, Tarakanov A, Goryanin I. Using medical microwave radiometry for brain temperature measurements. Drug Discov Today 2021; 27:881-889. [PMID: 34767961 DOI: 10.1016/j.drudis.2021.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 09/29/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022]
Abstract
Brain temperature (BT) is a crucial physiological parameter used to monitor cerebral status. Physical activities and traumatic brain injuries (TBI) can affect BT; therefore, non-invasive BT monitoring is an important way to gain insight into TBI, stroke, and wellbeing. The effects of BT on physical performance have been studied at length. When humans are under extreme conditions, most of the energy consumed is used to maintain the BT. In addition, measuring the BT is useful for early brain diagnostics. Passive microwave radiometry (MWR) measures the intrinsic radiation of tissues in the 1-4 GHz range. It was shown that non-invasive passive MWR technology can successfully measure BT and identify even small TBIs. Here, we review the potential applications of MWR for assessing BT.
Collapse
Affiliation(s)
- Oleg Shevelev
- People' Friendship University of Russia, Moscow, Russia; Federal Research and Clinical Centre for Resuscitation and Rehabilitation, Moscow, Russia
| | - Marina Petrova
- People' Friendship University of Russia, Moscow, Russia; Federal Research and Clinical Centre for Resuscitation and Rehabilitation, Moscow, Russia
| | - Andrey Smolensky
- Russian State University of Physical Culture, Sports, Youth and Tourism, Moscow, Russia
| | - Batyr Osmonov
- Educational - Scientifc Medical Center of Kyrgyz Medical Sate University, Bishkek, Kyrgyz Republic
| | | | - Tatyana Kharybina
- Library for Natural Sciences of the Russian Academy of Sciences, Moscow, Russia
| | | | | | - Sergey Vesnin
- Medical Microwave Radiometry Ltd, Edinburgh, UK; RTM Diagnostic LLC, Moscow, Russia; Bauman Moscow State Technical University, Moscow, Russia
| | | | - Igor Goryanin
- School of Informatics, University of Edinburgh, Edinburgh, UK; Institute Theoretical and Experimental Biophysics, Pushchino, Russia; Okinawa Institute Science and Technology, Okinawa, Japan.
| |
Collapse
|
11
|
Li Y, Ren M, Wang J, Ma R, Chen H, Xie Q, Li H, Li J, Wang J. Progress in Borneol Intervention for Ischemic Stroke: A Systematic Review. Front Pharmacol 2021; 12:606682. [PMID: 34017247 PMCID: PMC8129537 DOI: 10.3389/fphar.2021.606682] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Borneol is a terpene and bicyclic organic compound that can be extracted from plants or chemically synthesized. As an important component of proprietary Chinese medicine for the treatment of stroke, its neuroprotective effects have been confirmed in many experiments. Unfortunately, there is no systematic review of these studies. This study aimed to systematically examine the neuroprotective effects of borneol in the cascade reaction of experimental ischemic stroke at different periods. Methods: Articles on animal experiments and cell-based research on the actions of borneol against ischemic stroke in the past 20°years were collected from Google Scholar, Web of Science, PubMed, ScienceDirect, China National Knowledge Infrastructure (CNKI), and other biomedical databases. Meta-analysis was performed on key indicators in vivo experiments. After sorting the articles, we focused on the neuroprotective effects and mechanism of action of borneol at different stages of cerebral ischemia. Results: Borneol is effective in the prevention and treatment of nerve injury in ischemic stroke. Its mechanisms of action include improvement of cerebral blood flow, inhibition of neuronal excitotoxicity, blocking of Ca2+ overload, and resistance to reactive oxygen species injury in the acute ischemic stage. In the subacute ischemic stage, borneol may antagonize blood-brain barrier injury, intervene in inflammatory reactions, and prevent neuron excessive death. In the late stage, borneol promotes neurogenesis and angiogenesis in the treatment of ischemic stroke. Conclusion: Borneol prevents neuronal injury after cerebral ischemia via multiple action mechanisms, and it can mobilize endogenous nutritional factors to hasten repair and regeneration of brain tissue. Because the neuroprotective effects of borneol are mediated by various therapeutic factors, deficiency caused by a single-target drug is avoided. Besides, borneol promotes other drugs to pass through the blood-brain barrier to exert synergistic therapeutic effects.
Collapse
Affiliation(s)
- Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyan Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinxiu Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Kim B, Ahn JH, Kim DW, Lee TK, Kim YS, Shin MC, Cho JH, Kim YM, Park JH, Kang IJ, Lee JC, Won MH. Transient forebrain ischemia under hyperthermic condition accelerates memory impairment and neuronal death in the gerbil hippocampus by increasing NMDAR1 expression. Mol Med Rep 2021; 23:256. [PMID: 33537826 PMCID: PMC7893780 DOI: 10.3892/mmr.2021.11895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/30/2020] [Indexed: 12/22/2022] Open
Abstract
Altered expression levels of N‑methyl‑D‑aspartate receptor (NMDAR), a ligand‑gated ion channel, have a harmful effect on cellular survival. Hyperthermia is a proven risk factor of transient forebrain ischemia (tFI) and can cause extensive and severe brain damage associated with mortality. The objective of the present study was to investigate whether hyperthermic preconditioning affected NMDAR1 immunoreactivity associated with deterioration of neuronal function in the gerbil hippocampal CA1 region following tFI via histological and western blot analyses. Hyperthermic preconditioning was performed for 1 h before tFI, which was developed by ligating common carotid arteries for 5 min. tFI‑induced cognitive impairment under hyperthermia was worse compared with that under normothermia. Loss (death) of pyramidal neurons in the CA1 region occurred fast and was more severe under hyperthermia compared with that under normothermia. NMDAR1 immunoreactivity was not observed in the somata of pyramidal neurons of sham gerbils with normothermia. However, its immunoreactivity was strong in the somata and processes at 12 h post‑tFI. Thereafter, NMDAR1 immunoreactivity decreased with time after tFI. On the other hand, NMDAR1 immunoreactivity under hyperthermia was significantly increased in the somata and processes at 6 h post‑tFI. The change pattern of NMDAR1 immunoreactivity under hyperthermia was different from that under normothermia. Overall, accelerated tFI‑induced neuronal death under hyperthermia may be closely associated with altered NMDAR1 expression compared with that under normothermia.
Collapse
Affiliation(s)
- Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Yoon Sung Kim
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| |
Collapse
|
13
|
Ávila-Gómez P, Vieites-Prado A, Dopico-López A, Bashir S, Fernández-Susavila H, Gubern C, Pérez-Mato M, Correa-Paz C, Iglesias-Rey R, Sobrino T, Bustamante A, Wellmann S, Montaner J, Serena J, Castillo J, Hervella P, Campos F. Cold stress protein RBM3 responds to hypothermia and is associated with good stroke outcome. Brain Commun 2020; 2:fcaa078. [PMID: 33585816 PMCID: PMC7869850 DOI: 10.1093/braincomms/fcaa078] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
RNA-binding motif protein 3 is a molecular marker of hypothermia that has proved neuroprotective in neurodegenerative disease models. However, its relationship to the well-recognized therapeutic effect of hypothermia in ischaemic stroke had not been studied. In this work, the expression of RNA-binding motif protein 3 was investigated in ischaemic animal models subjected to systemic and focal brain hypothermia, specifically the effects of RNA-binding motif protein 3 silencing and overexpression on ischaemic lesions. Moreover, the association of RNA-binding motif protein 3 levels with body temperature and clinical outcome was evaluated in two independent cohorts of acute ischaemic stroke patients (n = 215); these levels were also determined in a third cohort of 31 patients derived from the phase III EuroHYP-1 trial of therapeutic cooling in ischaemic stroke. The preclinical data confirmed the increase of brain RNA-binding motif protein 3 levels in ischaemic animals subjected to systemic and focal hypothermia; this increase was selectively higher in the cooled hemisphere of animals undergoing focal brain hypothermia, thus confirming the direct effect of hypothermia on RNA-binding motif protein 3 expression, while RNA-binding motif protein 3 up-regulation in ischaemic brain regions led to functional recovery. Clinically, patients with body temperature <37.5°C in the first two cohorts had higher RNA-binding motif protein 3 values at 24 h and good outcome at 3 months post-ischaemic stroke, while RNA-binding motif protein 3 levels in the cooled third cohort tended to exceed those in placebo-treated patients. These results make RNA-binding motif protein 3 a molecular marker associated with the effect of hypothermia in ischaemic stroke and suggest its potential application as a promising protective target.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Alba Vieites-Prado
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Antonio Dopico-López
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Saima Bashir
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - Héctor Fernández-Susavila
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Carme Gubern
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - María Pérez-Mato
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain.,Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Hospital La Paz Institute for Health Research (IdiPAZ), Autonomous University of Madrid, 28046, Madrid, Spain
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Sven Wellmann
- Department of Neonatology, University Children's Hospital Regensburg (KUNO), University of Regensburg, 93049 Regensburg, Germany
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Joaquín Serena
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| |
Collapse
|
14
|
Hosseini M, Wilson RH, Crouzet C, Amirhekmat A, Wei KS, Akbari Y. Resuscitating the Globally Ischemic Brain: TTM and Beyond. Neurotherapeutics 2020; 17:539-562. [PMID: 32367476 PMCID: PMC7283450 DOI: 10.1007/s13311-020-00856-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrest (CA) afflicts ~ 550,000 people each year in the USA. A small fraction of CA sufferers survive with a majority of these survivors emerging in a comatose state. Many CA survivors suffer devastating global brain injury with some remaining indefinitely in a comatose state. The pathogenesis of global brain injury secondary to CA is complex. Mechanisms of CA-induced brain injury include ischemia, hypoxia, cytotoxicity, inflammation, and ultimately, irreversible neuronal damage. Due to this complexity, it is critical for clinicians to have access as early as possible to quantitative metrics for diagnosing injury severity, accurately predicting outcome, and informing patient care. Current recommendations involve using multiple modalities including clinical exam, electrophysiology, brain imaging, and molecular biomarkers. This multi-faceted approach is designed to improve prognostication to avoid "self-fulfilling" prophecy and early withdrawal of life-sustaining treatments. Incorporation of emerging dynamic monitoring tools such as diffuse optical technologies may provide improved diagnosis and early prognostication to better inform treatment. Currently, targeted temperature management (TTM) is the leading treatment, with the number of patients needed to treat being ~ 6 in order to improve outcome for one patient. Future avenues of treatment, which may potentially be combined with TTM, include pharmacotherapy, perfusion/oxygenation targets, and pre/postconditioning. In this review, we provide a bench to bedside approach to delineate the pathophysiology, prognostication methods, current targeted therapies, and future directions of research surrounding hypoxic-ischemic brain injury (HIBI) secondary to CA.
Collapse
Affiliation(s)
- Melika Hosseini
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Robert H Wilson
- Department of Neurology, School of Medicine, University of California, Irvine, USA
- Beckman Laser Institute, University of California, Irvine, USA
| | - Christian Crouzet
- Department of Neurology, School of Medicine, University of California, Irvine, USA
- Beckman Laser Institute, University of California, Irvine, USA
| | - Arya Amirhekmat
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Kevin S Wei
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Yama Akbari
- Department of Neurology, School of Medicine, University of California, Irvine, USA.
- Beckman Laser Institute, University of California, Irvine, USA.
| |
Collapse
|
15
|
Jackson TC, Kochanek PM. A New Vision for Therapeutic Hypothermia in the Era of Targeted Temperature Management: A Speculative Synthesis. Ther Hypothermia Temp Manag 2019; 9:13-47. [PMID: 30802174 PMCID: PMC6434603 DOI: 10.1089/ther.2019.0001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Three decades of animal studies have reproducibly shown that hypothermia is profoundly cerebroprotective during or after a central nervous system (CNS) insult. The success of hypothermia in preclinical acute brain injury has not only fostered continued interest in research on the classic secondary injury mechanisms that are prevented or blunted by hypothermia but has also sparked a surge of new interest in elucidating beneficial signaling molecules that are increased by cooling. Ironically, while research into cold-induced neuroprotection is enjoying newfound interest in chronic neurodegenerative disease, conversely, the scope of the utility of therapeutic hypothermia (TH) across the field of acute brain injury is somewhat controversial and remains to be fully defined. This has led to the era of Targeted Temperature Management, which emphasizes a wider range of temperatures (33–36°C) showing benefit in acute brain injury. In this comprehensive review, we focus on our current understandings of the novel neuroprotective mechanisms activated by TH, and discuss the critical importance of developmental age germane to its clinical efficacy. We review emerging data on four cold stress hormones and three cold shock proteins that have generated new interest in hypothermia in the field of CNS injury, to create a framework for new frontiers in TH research. We make the case that further elucidation of novel cold responsive pathways might lead to major breakthroughs in the treatment of acute brain injury, chronic neurological diseases, and have broad potential implications for medicines of the distant future, including scenarios such as the prevention of adverse effects of long-duration spaceflight, among others. Finally, we introduce several new phrases that readily summarize the essence of the major concepts outlined by this review—namely, Ultramild Hypothermia, the “Responsivity of Cold Stress Pathways,” and “Hypothermia in a Syringe.”
Collapse
Affiliation(s)
- Travis C Jackson
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Endothelial Protrusions in Junctional Integrity and Barrier Function. CURRENT TOPICS IN MEMBRANES 2018; 82:93-140. [PMID: 30360784 DOI: 10.1016/bs.ctm.2018.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endothelial cells of the microcirculation form a semi-permeable diffusion barrier between the blood and tissues. This permeability of the endothelium, particularly in the capillaries and postcapillary venules, is a normal physiological function needed for blood-tissue exchange in the microcirculation. During inflammation, microvascular permeability increases dramatically and can lead to tissue edema, which in turn can lead to dysfunction of tissues and organs. The molecular mechanisms that control the barrier function of endothelial cells have been under investigation for several decades and remain an important topic due to the potential for discovery of novel therapeutic strategies to reduce edema. This review highlights current knowledge of the cellular and molecular mechanisms that lead to endothelial hyperpermeability during inflammatory conditions associated with injury and disease. This includes a discussion of recent findings demonstrating temporal protrusions by endothelial cells that may contribute to intercellular junction integrity between endothelial cells and affect the diffusion distance for solutes via the paracellular pathway.
Collapse
|
17
|
de Jonge JC, Wallet J, van der Worp HB. Fever worsens outcomes in animal models of ischaemic stroke: A systematic review and meta-analysis. Eur Stroke J 2018; 4:29-38. [PMID: 31165092 PMCID: PMC6533866 DOI: 10.1177/2396987318776421] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/17/2018] [Indexed: 01/05/2023] Open
Abstract
Background Subfebrile temperatures and fever in the first days after stroke are associated with a greater risk of a poor outcome. If this relation is causal, prevention of hyperthermia may improve outcome. Causality can be tested in animal models. We therefore assessed the effects of hyperthermia on outcomes in animal models of ischaemic stroke and explored under which conditions prevention of hyperthermia could be most effective. Methods We performed a systematic review and meta-analysis of data from animal experiments testing the effect of spontaneous or induced hyperthermia on outcome after focal cerebral ischaemia. Our primary outcome measure was infarct size. Normalised mean differences were combined using the random effects model and stratified meta-analysis was used to explore the impact of study characteristics. Results We included 19 publications, reporting on 49 comparisons involving 603 animals. Overall, hyperthermia increased infarct size by 43.4% (95% confidence interval, 29.8-56.9%) and worsened neurobehavioral outcomes by 48.5% (17.2-79.8%). The increase in infarct size was larger with higher temperatures. Hyperthermia was most harmful if present for more than 2 h and when started at the time of artery occlusion rather than later. Conclusion Hyperthermia substantially increased infarct size in animal models of ischaemic stroke, suggesting that the relation between fever and poor outcome observed in patients is at least in part causal. These data provide support to trials testing the effect of the prevention of fever with antipyretic drugs in patients with acute stroke.
Collapse
Affiliation(s)
- Jeroen C de Jonge
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Justin Wallet
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - H Bart van der Worp
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
18
|
MRI and MR Spectroscopy Features of Heat Stroke: A Case Report. IRANIAN JOURNAL OF RADIOLOGY 2018. [DOI: 10.5812/iranjradiol.62386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
19
|
Császár N, Scholkmann F, Salari V, Szőke H, Bókkon I. Phosphene perception is due to the ultra-weak photon emission produced in various parts of the visual system: glutamate in the focus. Rev Neurosci 2018; 27:291-9. [PMID: 26544101 DOI: 10.1515/revneuro-2015-0039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/11/2015] [Indexed: 12/24/2022]
Abstract
Phosphenes are experienced sensations of light, when there is no light causing them. The physiological processes underlying this phenomenon are still not well understood. Previously, we proposed a novel biopsychophysical approach concerning the cause of phosphenes based on the assumption that cellular endogenous ultra-weak photon emission (UPE) is the biophysical cause leading to the sensation of phosphenes. Briefly summarized, the visual sensation of light (phosphenes) is likely to be due to the inherent perception of UPE of cells in the visual system. If the intensity of spontaneous or induced photon emission of cells in the visual system exceeds a distinct threshold, it is hypothesized that it can become a conscious light sensation. Discussing several new and previous experiments, we point out that the UPE theory of phosphenes should be really considered as a scientifically appropriate and provable mechanism to explain the physiological basis of phosphenes. In the present paper, we also present our idea that some experiments may support that the cortical phosphene lights are due to the glutamate-related excess UPE in the occipital cortex.
Collapse
|
20
|
Naderi S, Alimohammadi R, Hakimizadeh E, Roohbakhsh A, Shamsizadeh A, Allahtavakoli M. The effect of exercise preconditioning on stroke outcome in ovariectomized mice with permanent middle cerebral artery occlusion. Can J Physiol Pharmacol 2017; 96:287-294. [PMID: 28873322 DOI: 10.1139/cjpp-2017-0157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exercise preconditioning has been shown to be effective in improving behavioral and neuropathological indices after cerebral ischemia. We evaluated the effect of exercise preconditioning, 17β-estradiol, and their combination on stroke outcome using an experimental model of stroke in ovariectomized (OVX) mice. OVX mice were randomly assigned to 4 groups as follows: control (stroke), exercise (exercise and stroke), estradiol (17β-estradiol and stroke), and exercise+estradiol (exercise and 17β-estradiol and stroke). Exercise preconditioning was performed on a treadmill 5 days/week, 40 min/day, at a speed of 18 m/min for 4 weeks. 17β-estradiol was gavaged (40 μg/kg per day) for 4 weeks. Stroke was induced by permanent middle cerebral artery occlusion (pMCAO), and neurological deficits were evaluated 1, 2, and 7 days after stroke. Then, the serum concentrations of matrix metalloproteinase-9 (MMP-9) and interleukin-10 (IL-10) and infarct volumes were assessed. Exercise preconditioning and 17β-estradiol induced a better outcome compared with the control ischemic mice, which was manifested by decrease in MMP-9, increase in IL-10, diminished infarct volume, and improved neurological deficits. Concomitant administration of 17β-estradiol and exercise also significantly improved these parameters. Exercise preconditioning or administration of 17β-estradiol alone or in combination before pMCAO induced significant neuroprotection in OVX mice.
Collapse
Affiliation(s)
- Soudabeh Naderi
- a Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Raheleh Alimohammadi
- a Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Hakimizadeh
- d Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Roohbakhsh
- b Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,c Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shamsizadeh
- d Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- d Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
21
|
Lee JH, Zhang J, Yu SP. Neuroprotective mechanisms and translational potential of therapeutic hypothermia in the treatment of ischemic stroke. Neural Regen Res 2017; 12:341-350. [PMID: 28469636 PMCID: PMC5399699 DOI: 10.4103/1673-5374.202915] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Stroke is a leading cause of disability and death, yet effective treatments for acute stroke has been very limited. Thus far, tissue plasminogen activator has been the only FDA-approved drug for thrombolytic treatment of ischemic stroke patients, yet its application is only applicable to less than 4–5% of stroke patients due to the narrow therapeutic window (< 4.5 hours after the onset of stroke) and the high risk of hemorrhagic transformation. Emerging evidence from basic and clinical studies has shown that therapeutic hypothermia, also known as targeted temperature management, can be a promising therapy for patients with different types of stroke. Moreover, the success in animal models using pharmacologically induced hypothermia (PIH) has gained increasing momentum for clinical translation of hypothermic therapy. This review provides an updated overview of the mechanisms and protective effects of therapeutic hypothermia, as well as the recent development and findings behind PIH treatment. It is expected that a safe and effective hypothermic therapy has a high translational potential for clinical treatment of patients with stroke and other CNS injuries.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| | - James Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| |
Collapse
|
22
|
Vieites-Prado A, Iglesias-Rey R, Fernández-Susavila H, da Silva-Candal A, Rodríguez-Castro E, Gröhn OHJ, Wellmann S, Sobrino T, Castillo J, Campos F. Protective Effects and Magnetic Resonance Imaging Temperature Mapping of Systemic and Focal Hypothermia in Cerebral Ischemia. Stroke 2016; 47:2386-96. [PMID: 27491739 DOI: 10.1161/strokeaha.116.014067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/30/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Hypothermia is potentially the most effective protective therapy for brain ischemia; however, its use is limited because of serious side effects. Although focal hypothermia (FH) has a significantly lower stress profile than systemic hypothermia (SH), its efficacy in ischemia has been poorly studied. We aimed to compare the therapeutic effects of each treatment on various short- and long-term clinically relevant end points. METHODS Sprague-Dawley rats were subjected to transient (45 minutes) occlusion of the middle cerebral artery. One hour after arterial reperfusion, animals were randomly assigned to groups for treatment with SH or FH (target temperature: 32°C) for 4 or 24 hours. Lesion volume, edema, functional recovery, and histological markers of cellular injury were evaluated for 1 month after ischemic injury. Effects of SH and FH on cerebral temperature were also analyzed for the first time by magnetic resonance thermometry, an approach that combines spectroscopy with gradient-echo-based phase mapping. RESULTS Both therapeutic approaches reduced ischemic lesion volume (P<0.001), although a longer FH treatment (24 hours) was required to achieve similar protective effects to those induced by 4 hours of SH. In addition, magnetic resonance thermometry demonstrated that systemic hypothermia reduced whole-brain temperature, whereas FH primarily reduced the temperature of the ischemic region. CONCLUSIONS Focal brain hypothermia requires longer cooling periods to achieve the same protective efficacy as SH. However, FH mainly affects the ischemic region, and therefore represents a promising and nonstressful alternative to SH.
Collapse
Affiliation(s)
- Alba Vieites-Prado
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Ramón Iglesias-Rey
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Héctor Fernández-Susavila
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Andrés da Silva-Candal
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Emilio Rodríguez-Castro
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Olli H J Gröhn
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Sven Wellmann
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - Tomás Sobrino
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.)
| | - José Castillo
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.).
| | - Francisco Campos
- From the Clinical Neurosciences Research Laboratory, Clinical University Hospital, Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain (A.V.-P., R.I.-R., H.F.-S., A.d.S.-C., E.R.-C., T.S., J.C., F.C.); Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (O.H.J.G.); and Division of Neonatology, University of Basel Children's Hospital (UKBB), Switzerland (S.W.).
| |
Collapse
|
23
|
|
24
|
Salari V, Scholkmann F, Bokkon I, Shahbazi F, Tuszynski J. The Physical Mechanism for Retinal Discrete Dark Noise: Thermal Activation or Cellular Ultraweak Photon Emission? PLoS One 2016; 11:e0148336. [PMID: 26950936 PMCID: PMC4780715 DOI: 10.1371/journal.pone.0148336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/15/2016] [Indexed: 01/05/2023] Open
Abstract
For several decades the physical mechanism underlying discrete dark noise of photoreceptors in the eye has remained highly controversial and poorly understood. It is known that the Arrhenius equation, which is based on the Boltzmann distribution for thermal activation, can model only a part (e.g. half of the activation energy) of the retinal dark noise experimentally observed for vertebrate rod and cone pigments. Using the Hinshelwood distribution instead of the Boltzmann distribution in the Arrhenius equation has been proposed as a solution to the problem. Here, we show that the using the Hinshelwood distribution does not solve the problem completely. As the discrete components of noise are indistinguishable in shape and duration from those produced by real photon induced photo-isomerization, the retinal discrete dark noise is most likely due to ‘internal photons’ inside cells and not due to thermal activation of visual pigments. Indeed, all living cells exhibit spontaneous ultraweak photon emission (UPE), mainly in the optical wavelength range, i.e., 350–700 nm. We show here that the retinal discrete dark noise has a similar rate as UPE and therefore dark noise is most likely due to spontaneous cellular UPE and not due to thermal activation.
Collapse
Affiliation(s)
- Vahid Salari
- Department of Physics, Isfahan University of Technology, Isfahan 84156-83111, Iran
- School of Physics, Institute for Research in Fundamental Sciences (IPM), Tehran 19395-5531, Iran
| | - Felix Scholkmann
- Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Research Office for Complex Physical and Biological Systems (ROCoS), 8038 Zurich, Switzerland
| | - Istvan Bokkon
- Vision Research Institute, 25 Rita Street, Lowell, MA 01854, United States of America
- Psychoszomatic OutPatient Department of the National Center for Spinal Disorders, Budapest H-1126, Hungary
| | - Farhad Shahbazi
- Department of Physics, Isfahan University of Technology, Isfahan 84156-83111, Iran
- School of Physics, Institute for Research in Fundamental Sciences (IPM), Tehran 19395-5531, Iran
| | - Jack Tuszynski
- Department of Physics, University of Alberta, T6G 2J1, Edmonton, AB, Canada
- * E-mail:
| |
Collapse
|
25
|
Ji C, Wang L, Dai R, Shan L, Yang H, Zhu H, Meng Q. Hyperthermia exacerbates the effects of cathepsin L on claudin-1 in a blood-brain barrier model in vitro. Brain Res 2015; 1631:72-9. [PMID: 26655064 DOI: 10.1016/j.brainres.2015.11.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 01/17/2023]
Abstract
PURPOSE The effects of cathepsin L on claudin-1 expression were investigated under hyperthermic condition in a blood-brain barrier (BBB) model in vitro, in order to estimate the potential effects of hyperthermia on BBB dysfunction. MATERIALS AND METHODS Brain microvascular endothelial cells (BMECs) and astrocytes were obtained from rat brain. The BBB models were randomly divided into a sham (37°C) group, a 39°C group, a 37°C+cathepsin L group and a 39°C+cathepsin L group. The permeability of BBB was judged. The expressions of cathepsin L in astrocytes and claudin-1 in BMECs were detected using immunohistochemistry method and western blot assay. RESULTS The permeability of BBB models was higher in the 39°C group than in the sham group. The cathepsin L expression in astrocytes was higher in the 39°C group than in the sham group (P<0.01), whereas the claudin-1 expression in BMECs was lower in the 39°C group than in the sham group (P<0.01). The claudin-1 expression in BMECs was significantly lower in the 37°C+cathepsin L group than in the sham group (P<0.01). At the same time point, the claudin-1 expression in BMECs was significantly lower in the 39°C+cathepsin L group than in the 37°C+cathepsin L group (P<0.01). CONCLUSION Hyperthermia can probably decrease claudin-1 expression in BMECs by upregulating cathepsin L expression in astrocytes in a BBB model in vitro.
Collapse
Affiliation(s)
- Conghua Ji
- Department of Postgraduate, Kunming Medical University, Kunming, Yunnan, China
| | - Limin Wang
- Department of Postgraduate, Kunming Medical University, Kunming, Yunnan, China
| | - Rongrong Dai
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li Shan
- Department of Postgraduate, Kunming Medical University, Kunming, Yunnan, China
| | - Hui Yang
- Department of Pathology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Hongyan Zhu
- Department of Clinical Laboratory, the Affiliated Kunhua Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiang Meng
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| |
Collapse
|
26
|
Yan G, Xuan Y, Dai Z, Zhang G, Xu H, Mikulis D, Wu R. Evolution of blood-brain barrier damage associated with changes in brain metabolites following acute ischemia. Neuroreport 2015; 26:945-51. [PMID: 26366833 DOI: 10.1097/wnr.0000000000000438] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Stroke is a serious medical condition that requires emergency care. In the case of ischemic stroke, ischemia may lead to damage to the blood-brain barrier (BBB); the damage in turn may exacerbate the condition. Therefore, noninvasive detection of BBB damage represents a challenge for experimental and clinical researchers. In this study, we assessed the onset of BBB disruption by means of T1-weighted images with administration of the contrast enhancement agent gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA) and related BBB breakdown to brain metabolite changes in proton magnetic resonance spectrum (H-MRS) in the infarcted site following middle cerebral artery occlusion (MCAO) in rats. It was shown that MCAO for 30 min and 1.5 h caused no Gd-DTPA signal change in the T1-weighted images, whereas MCAO for 1 h significantly altered some of H-MRS brain metabolites, suggesting that brain metabolite changes occurred earlier than BBB damage after ischemic stroke. MCAO for 2 h caused BBB breakdown, which was related to changes in the levels of some brain metabolites detected by H-MRS. Between the second and the third hour after MCAO, brain metabolite changes continued as the result of BBB breakdown and the concurrent overperfusion to the infarcted site, which may ameliorate the metabolite changes, thus compensating for the functional failures of the brain after stroke.
Collapse
Affiliation(s)
- Gen Yan
- aDepartment of Radiology, Affiliated Hospital, Jiangnan University bDepartment of Basic Medicine, Jiangnan University Medical School, Wuxi cDepartment of Radiology, the Second Affiliated Hospital, Shantou University Medical College, Shantou dDepartment of Anatomy, Shantou University Medical College, Shantou, People's Republic of China eDepartment of Medical Imaging, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | |
Collapse
|
27
|
Otto KA. Therapeutic hypothermia applicable to cardiac surgery. Vet Anaesth Analg 2015; 42:559-69. [PMID: 26361886 DOI: 10.1111/vaa.12299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/19/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To review the beneficial and adverse effects of therapeutic hypothermia (TH) applicable to cardiac surgery with cardiopulmonary bypass (CPB) in the contexts of various temperature levels and techniques for achieving TH. DATABASES USED Multiple electronic literature searches were performed using PubMed and Google for articles published from June 2012 to December 2014. Relevant terms (e.g. 'hypothermia', 'cardiopulmonary bypass', 'cardiac surgery', 'neuroprotection') were used to search for original articles, letters and reviews without species limitation. Reviews were included despite potential publication bias. References from the studies identified were also searched to find other potentially relevant citations. Abstracts, case reports, conference presentations, editorials and expert opinions were excluded. CONCLUSIONS Therapeutic hypothermia is an essential measure of neuroprotection during cardiac surgery that may be achieved most effectively by intravascular cooling using hypothermic CPB. For most cardiac surgical procedures, mild to modest (32-36 °C) TH will be sufficient to assure neuroprotection and will avoid most of the adverse effects of hypothermia that occur at lower body core temperatures.
Collapse
Affiliation(s)
- Klaus A Otto
- Central Laboratory Animal Facility, Hannover Medical School, Hannover, Germany
| |
Collapse
|
28
|
Kersemans V, Gilchrist S, Allen PD, Beech JS, Kinchesh P, Vojnovic B, Smart SC. A resistive heating system for homeothermic maintenance in small animals. Magn Reson Imaging 2015; 33:847-51. [PMID: 25863135 PMCID: PMC4462590 DOI: 10.1016/j.mri.2015.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/12/2015] [Accepted: 03/30/2015] [Indexed: 01/27/2023]
Abstract
PURPOSE To develop an MR-compatible resistive heater for temperature maintenance of anaesthetized animals. MATERIALS AND METHODS An MR-compatible resistive electrical heater was formed from a tightly-wound twisted pair wire, interfaced to a homeothermic maintenance controller. Fat-suppressed images and localized spectra were acquired with the twisted pair heater and a near-identical single strand heater during operation at maximum power. Data were also acquired in the absence of heating to demonstrate the insensitivity of MR to distortions arising from the passage of current through the heater elements. The efficacy of temperature maintenance was examined by measuring rectal temperature immediately following induction of general anesthesia and throughout and after the acquisition of a heater artifact-prone image series. RESULTS Images and spectra acquired in the presence and absence of DC current through the twisted pair heater were identical whereas the passage of current through the single strand wire created field shifts and lineshape distortions. Temperature that is lost during anesthesia induction was recovered within approximately 10-20 minutes of induction, and a stable temperature is reached as the animal's temperature approaches the set target. CONCLUSION The twisted pair wire heater does not interfere with MR image quality and maintains adequate thermal input to the animal to maintain body temperature.
Collapse
Affiliation(s)
- Veerle Kersemans
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| | - Stuart Gilchrist
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| | - Philip D Allen
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| | - John S Beech
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| | - Paul Kinchesh
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| | - Borivoj Vojnovic
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| | - Sean C Smart
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, OX3-7DQ, Oxford, United Kingdom.
| |
Collapse
|
29
|
Ruban A, Malina KCK, Cooper I, Graubardt N, Babakin L, Jona G, Teichberg VI. Combined Treatment of an Amyotrophic Lateral Sclerosis Rat Model with Recombinant GOT1 and Oxaloacetic Acid: A Novel Neuroprotective Treatment. NEURODEGENER DIS 2015; 15:233-42. [PMID: 26113413 DOI: 10.1159/000382034] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/02/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIM The sporadic form of the disease affects the majority of amyotrophic lateral sclerosis (ALS) patients. The role of glutamate (Glu) excitotoxicity in ALS has been extensively documented and remains one of the prominent hypotheses of ALS pathogenesis. In light of this evidence, the availability of a method to remove excess Glu from brain and spinal cord extracellular fluids without the need to deliver drugs across the blood-brain barrier and with minimal or no adverse effects may provide a major therapeutic asset, which is the primary aim of this study. METHODS The therapeutic efficacy of the combined treatment with recombinant Glu-oxaloacetate-transaminase (rGOT) and its co-factor oxaloacetic acid (OxAc) has been tested in an animal model of sporadic ALS. RESULTS We found that OxAc/rGOT treatment provides significant neuroprotection to spinal cord motor neurons. It also slows down the development of motor weakness and prolongs survival. CONCLUSION In this study we bring evidence that the administration of Glu scavengers to rats with sporadic ALS inhibited the massive death of spinal cord motor neurons, slowed the onset of motor weakness and prolonged survival. This treatment may be of high clinical significance for the future treatment of chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Angela Ruban
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | |
Collapse
|
30
|
Nishiyama M, Nagase H, Tanaka T, Fujita K, Maruyama A, Toyoshima D, Nakagawa T, Taniguchi-Ikeda M, Morioka I, Morisada N, Takada S, Iijima K. Demographics and outcomes of patients with pediatric febrile convulsive status epilepticus. Pediatr Neurol 2015; 52:499-503. [PMID: 25769239 DOI: 10.1016/j.pediatrneurol.2015.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Convulsive status epilepticus with fever is common and may be related to neurological sequela in children. However, there are limited data on the demographics and risk factors of this phenomenon. Thus, we aimed to describe the demographics and risk factors of neurological sequela among children with convulsive status epilepticus with fever. METHODS We reviewed convulsive status epilepticus with fever cases in the pediatric intensive care unit at Kobe Children's Hospital between 2002 and 2013. We included patients with intrinsic neurological disease, and excluded those with obvious central nervous system infection. Cases of neurological worsening were categorized as poor outcome using the pediatric cerebral performance category scale. Possible risk factors for poor outcome included age, sex, neurological medical history, seizure duration, body temperature, and level of consciousness. RESULTS A total of 253 patients (128 males), aged 1 month to 15 years (mean 45 ± 40 months), were enrolled. Three patients (1.2%) died during hospitalization, and 32 (12.6%) patients had a poor outcome. A univariate analysis identified male sex, absence of epilepsy history, body temperature above 40°C on admission, seizure duration longer than 120 minutes, impaired consciousness at 12 hours after onset, and presence of nonconvulsive seizure as potential predictors of poor outcome. A multivariate analysis, revealed that an absence of epilepsy history (odds ratio = 11.18), body temperature above 40°C on admission (odds ratio = 3.39), or impaired consciousness at 12 hours after onset (odds ratio = 41.85) was associated with poor outcome. CONCLUSIONS Our study indicated that absence of epilepsy history, high temperature, and/or prolonged impaired consciousness were associated with brain injury.
Collapse
Affiliation(s)
- Masahiro Nishiyama
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Neurology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan.
| | - Hiroaki Nagase
- Department of Neurology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Tsukasa Tanaka
- Department of Neurology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Kyoko Fujita
- Department of Neurology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Azusa Maruyama
- Department of Neurology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Daisaku Toyoshima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taku Nakagawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - Ichiro Morioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoya Morisada
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Takada
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
31
|
Neuroprotection for ischaemic stroke: Current status and challenges. Pharmacol Ther 2015; 146:23-34. [DOI: 10.1016/j.pharmthera.2014.09.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/31/2022]
|
32
|
Hu W, Cheng X, Ye X, Zhao L, Huang Y, Zhu H, Yan Z, Wang X, Wang X, Bai G, Gao H. Ex vivo (1)H nuclear magnetic resonance spectroscopy reveals systematic alterations in cerebral metabolites as the key pathogenetic mechanism of bilirubin encephalopathy. Mol Brain 2014; 7:87. [PMID: 25424547 PMCID: PMC4252999 DOI: 10.1186/s13041-014-0087-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/13/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bilirubin encephalopathy (BE) is a severe neurologic sequelae induced by hyperbilirubinemia in newborns. However, the pathogenetic mechanisms underlying the clinical syndromes of BE remain ambiguous. Ex vivo (1)H nuclear magnetic resonance (NMR) spectroscopy was used to measure changes in the concentrations of cerebral metabolites in various brain areas of newborn 9-day-old rats subjected to bilirubin to explore the related mechanisms of BE. RESULTS When measured 0.5 hr after injection of bilirubin, levels of the amino acid neurotransmitters glutamate (Glu), glutamine (Gln), and γ-aminobutyric acid (GABA) in hippocampus and occipital cortex significantly decreased, by contrast, levels of aspartate (Asp) considerably increased. In the cerebellum, Glu and Gln levels significantly decreased, while GABA, and Asp levels showed no significant differences. In BE 24 hr rats, all of the metabolic changes observed returned to normal in the hippocampus and occipital cortex; however, levels of Glu, Gln, GABA, and glycine significantly increased in the cerebellum. CONCLUSIONS These metabolic changes for the neurotransmitters are mostly likely the result of a shift in the steady-state equilibrium of the Gln-Glu-GABA metabolic cycle between astrocytes and neurons, in a region-specific manner. Changes in energy metabolism and the tricarboxylic acid cycle may also be involved in the pathogenesis of BE.
Collapse
Affiliation(s)
- Wenyi Hu
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiaojie Cheng
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Xinjian Ye
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Liangcai Zhao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Yanan Huang
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Huanle Zhu
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Zhihan Yan
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Xuebao Wang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiaojie Wang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Guanghui Bai
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
33
|
Xu J, Qian Z, Zhao L, Fang Y. An optimized HPLC/MS/MS method for quantification of excitatory amino acids in rat hippocampus and its application in brain ischemia/reperfusion research. Biomed Chromatogr 2014; 28:1822-7. [PMID: 24816475 DOI: 10.1002/bmc.3226] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/20/2014] [Accepted: 04/03/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Jia Xu
- Department of Pharmacy; Mawangdui Hospital; Changsha 410016 China
| | - Zhonglian Qian
- Department of Pharmacy; The First Hospital; Huhhot 010010 China
| | - Libo Zhao
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
| | - Yi Fang
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
| |
Collapse
|
34
|
Boyko M, Gruenbaum SE, Gruenbaum BF, Shapira Y, Zlotnik A. Brain to blood glutamate scavenging as a novel therapeutic modality: a review. J Neural Transm (Vienna) 2014; 121:971-9. [PMID: 24623040 DOI: 10.1007/s00702-014-1181-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 02/11/2014] [Indexed: 12/27/2022]
Abstract
It is well known that abnormally elevated glutamate levels in the brain are associated with secondary brain injury following acute and chronic brain insults. As such, a tight regulation of brain glutamate concentrations is of utmost importance in preventing the neurodegenerative effects of excess glutamate. There has been much effort in recent years to better understand the mechanisms by which glutamate is reduced in the brain to non-toxic concentrations, and in how to safely accelerate these mechanisms. Blood glutamate scavengers such as oxaloacetate, pyruvate, glutamate-oxaloacetate transaminase, and glutamate-pyruvate transaminase have been shown to reduce blood glutamate concentrations, thereby increasing the driving force of the brain to blood glutamate efflux and subsequently reducing brain glutamate levels. In the past decade, blood glutamate scavengers have gained increasing international interest, and its uses have been applied to a wide range of experimental contexts in animal models of traumatic brain injury, ischemic stroke, subarachnoid hemorrhage, epilepsy, migraine, and malignant gliomas. Although glutamate scavengers have not yet been used in humans, there is increasing evidence that their use may provide effective and exciting new therapeutic modalities. Here, we review the laboratory evidence for the use of blood glutamate scavengers. Other experimental neuroprotective treatments thought to scavenge blood glutamate, including estrogen and progesterone, beta-adrenergic activation, hypothermia, insulin and glucagon, and hemodialysis and peritoneal dialysis are also discussed. The evidence reviewed here will hopefully pave the way for future clinical trials.
Collapse
Affiliation(s)
- Matthew Boyko
- Department of Anesthesiology and Critical Care, Faculty of Health Sciences, Soroka Medical Center Ben Gurion University of the Negev, Beer Sheba, Israel
| | | | | | | | | |
Collapse
|
35
|
Hyperthermia in human ischemic and hemorrhagic stroke: similar outcome, different mechanisms. PLoS One 2013; 8:e78429. [PMID: 24223804 PMCID: PMC3817202 DOI: 10.1371/journal.pone.0078429] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/19/2013] [Indexed: 12/20/2022] Open
Abstract
Hyperthermia is a predictor of poor outcome in ischemic (IS) and intracerebral hemorrhagic (ICH) stroke. Our aim was to study the plausible mechanisms involved in the poor outcome associated to hyperthermia in stroke. We conducted a case-control study including patients with IS (n = 100) and ICH (n = 100) within the first 12 hours from symptom onset. Specifically, IS and ICH patients were consecutively included into 2 subgroups, according to the highest body temperature within the first 24 hours: Tmax <37.5°C and Tmax ≥37.5°C, up to reach 50 patients per subgroup of temperature for both IS and ICH patients. Body temperature was determined at admission and every 4 hours during the first 48 hours. Main outcome variable was poor functional outcome (modified Rankin scale score >2) at 3 months. Serum levels of glutamate and active MMP-9 were measured at admission. Our results showed that Tmax ≥37.5°C within the first 24 hours was independently associated with poor outcome in both IS (OR, 12.43; 95% CI, 3.73-41.48; p<0.0001) and ICH (OR, 4.29; 95% CI, 1.32-13.91; p = 0.015) after adjusting for variables with a proven biological relevance for outcome. However, when molecular markers levels were included in the logistic regression model, we observed that glutamate (OR, 1.01; 95% CI, 1.00-1.02; p = 0.001) and infarct volume (OR, 1.06; 95% CI, 1.01-1.10; p = 0.015) were the only variables independently associated to poor outcome in IS, and active MMP-9 (OR, 1.04; 95% CI, 1.00-1.08; p = 0.002) and National Institute of Health Stroke Scale (NIHSS) at admission (OR, 1.29; 95% CI, 1.13-1.49; p<0.0001) in ICH. In conclusion, these results suggest that although the outcome associated to hyperthermia is similar in human IS and ICH, the underlying mechanisms may be different.
Collapse
|
36
|
Murase K, Assanai P, Takata H, Saito S, Nishiura M. A simple and inexpensive system for controlling body temperature in small animal experiments using MRI and the effect of body temperature on the hepatic kinetics of Gd-EOB-DTPA. Magn Reson Imaging 2013; 31:1744-51. [PMID: 24094686 DOI: 10.1016/j.mri.2013.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/28/2013] [Accepted: 08/20/2013] [Indexed: 01/31/2023]
Abstract
The purpose of this study was to develop a simple and inexpensive system for controlling body temperature in small animal experiments using magnetic resonance imaging (MRI) and to investigate the effect of body temperature on the kinetic behavior of gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid (Gd-EOB-DTPA) in the liver. In our temperature-control system, body temperature was controlled using a feedback-regulated heated or cooled air flow generated by two Futon dryers. The switches of the two Futon dryers were controlled using a digital temperature controller, in which the rectal temperature of a mouse measured by an optical fiber thermometer was used as the input. In experimental studies, male ICR mice aged 8weeks old were used and allocated into 5 groups (39-, 36-, 33-, 30-, and 27-degree groups, n=10), in which the body temperature was maintained at 39 °C, 36 °C, 33 °C, 30 °C, and 27 °C, respectively, using our system. The dynamic contrast-enhanced MRI (DCE-MRI) data were acquired with an MRI system for animal experiments equipped with a 1.5-Tesla permanent magnet, for approximately 43min, after the injection of Gd-EOB-DTPA into the tail vein. After correction of the image shift due to the temperature-dependent drift of the Larmor frequency using the gradient-based image registration method with robust estimation of displacement parameters, the kinetic behavior of Gd-EOB-DTPA was analyzed using an empirical mathematical model. With the use of this approach, the upper limit of the relative enhancement (A), the rates of contrast uptake (α) and washout (β), the parameter related to the slope of early uptake (q), the area under the curve (AUC), the maximum relative enhancement (REmax), the time to REmax (Tmax), and the elimination half-life of the contrast agent (T1/2) were calculated. The body temperature of mice could be controlled well by use of our system. Although there were no significant differences in α, AUC, and q among groups, there were significant differences in A, REmax, β, Tmax, and T1/2, indicating that body temperature significantly affects the kinetic behavior of Gd-EOB-DTPA in the liver. In conclusion, our system will be useful for controlling body temperature in small animal experiments using MRI. Because body temperature significantly affects the kinetic behavior of Gd-EOB-DTPA in the liver, the control of body temperature is essential and should be carefully considered when performing DCE-MRI studies in small animal experiments.
Collapse
Affiliation(s)
- Kenya Murase
- Department of Medical Physics and Engineering, Division of Medical Technology and Science, Faculty of Health Science, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | |
Collapse
|
37
|
Spielvogel D, Tang GHL. Selective cerebral perfusion for cerebral protection: what we do know. Ann Cardiothorac Surg 2013; 2:326-30. [PMID: 23977601 DOI: 10.3978/j.issn.2225-319x.2013.03.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 03/06/2013] [Indexed: 01/05/2023]
Abstract
Selective antegrade cerebral perfusion (SACP) for aortic arch surgery has evolved considerably since it was first reported. Various pressure rates have been investigated through animal models, as has the effect of warmer perfusate temperatures and hematocrit. Clinical research into pH management, the role of unilateral and bilateral perfusion, and core temperatures have further refined the procedure. We recommend the following protocol for SACP: perfusion pressure between 40-60 mmHg, flow rates between 6-10 mL/kg/min, and perfusate temperature of 20-28 °C; core cooling to 18-30 °C contingent on duration of arrest; alpha-stat pH management; hematocrit between 25-30%; near infrared spectroscopy to monitor cerebral perfusion; and bilateral perfusion when prolonged durations of SACP is anticipated.
Collapse
Affiliation(s)
- David Spielvogel
- Department of Surgery, Section of Cardiothoracic Surgery, New York Medical College, Westchester Medical Center, Valhalla, New York, USA
| | | |
Collapse
|
38
|
The role of hypothermia in the regulation of blood glutamate levels in naive rats. J Neurosurg Anesthesiol 2013; 25:174-83. [PMID: 23295267 DOI: 10.1097/ana.0b013e31827ee0ac] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The exact mechanism of hypothermia-induced neuroprotection has not been determined yet; however, we hypothesized that it may be mediated by a blood glutamate-scavenging effect. Here, we examine the effect of hypothermic conditions (mild, moderate, and deep) on blood glutamate levels in naive rats. To identify the mechanism of hypothermia-induced glutamate reduction, we also measured concentrations of glutamate oxaloacetate transaminase (GOT) and glutamate pyruvate transaminase (GPT), the primary regulators of glutamate concentration in blood. METHODS Rats were anesthetized with isoflurane, and their rectal temperature was maintained for 6 hours at 36 to 37°C, 33 to 36°C, 30 to 32°C, 18 to 22°C, or was not maintained artificially. At 6 hours, active cooling was discontinued and rats were allowed to rewarm. There were 12 rats in each group for a total of 60 rats. Blood samples were drawn at 0, 3, 6, 12, 24, and 48 hours for the determination of blood glutamate, GOT, and GPT levels. RESULTS A strong correlation between body temperature and blood glutamate levels was observed (P<0.001). Mild (33 to 36°C) and moderate (30 to 32°C) hypothermia led to reduced blood glutamate levels (P<0.001). Deep hypothermia (18 to 22°C) was associated with significant elevations in blood glutamate levels (P<0.001). Hypothermia, irrespective of the degree, led to elevations in GOT in plasma (P<0.001). CONCLUSIONS Mild and moderate hypothermia led to a reduction in blood glutamate levels in rats, whereas deep hypothermia was associated with a significant elevation in blood glutamate levels. We further demonstrated an elevation of GOT and GPT levels, supporting their involvement in reducing blood glutamate by the conversion of glutamate to 2-ketoglutarate. We suggest that the neuroprotective properties of hypothermia may be partially because of a blood glutamate-scavenging mechanism.
Collapse
|
39
|
Liu X, Wang M, Chen H, Guo Y, Ma F, Shi F, Bi Y, Li Y. Hypothermia protects the brain from transient global ischemia/reperfusion by attenuating endoplasmic reticulum response-induced apoptosis through CHOP. PLoS One 2013; 8:e53431. [PMID: 23301071 PMCID: PMC3536702 DOI: 10.1371/journal.pone.0053431] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/28/2012] [Indexed: 01/09/2023] Open
Abstract
Endoplasmic reticulum (ER) stress has been implicated in the pathology of cerebral ischemia. Apoptotic cell death occurs during prolonged period of stress or when the adaptive response fails. Hypothermia blocked the TNF or Fas-mediated extrinsic apoptosis pathway and the mitochondria pathway of apoptosis, however, whether hypothermia can block endoplasmic reticulum mediated apoptosis is never known. This study aimed to elucidate whether hypothermia attenuates brain cerebral ischemia/reperfusion (I/R) damage by suppressing ER stress-induced apoptosis. A 15 min global cerebral ischemia rat model was used in this study. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) positive cells in hippocampus CA1 were assessed after reperfusion of the brain. The expressions of C/EBP-homologous protein (CHOP) and glucose-regulated protein 78 (GRP78) in ischemic hippocampus CA1 were measured at 6, 12, 24 and 48 h after reperfusion. The results showed that hypothermia significantly attenuated brain I/R injury, as shown by reduction in cell apoptosis, CHOP expression, and increase in GRP78 expression. These results suggest that hypothermia could protect brain from I/R injury by suppressing ER stress-induced apoptosis.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Mingshan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Huailong Chen
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Yunliang Guo
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Fuguo Ma
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Fei Shi
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| | - Ying Li
- Department of Anesthesiology, Qingdao Municipal Hospital, Shandong, China
| |
Collapse
|
40
|
Zhao J, Li YX, Hao YJ, Chen R, Zhang JZ, Sun T, Yu JQ. Effects of oxysophoridine on rat hippocampal neurons sustained oxygen-glucose deprivation and reperfusion. CNS Neurosci Ther 2012; 19:138-41. [PMID: 23279847 DOI: 10.1111/cns.12047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 11/07/2012] [Accepted: 11/13/2012] [Indexed: 10/27/2022] Open
|