1
|
Alvarez FJ, Alvarez AA, Rodríguez JJ, Lafuente H, Canduela MJ, Hind W, Blanco-Bruned JL, Alonso-Alconada D, Hilario E. Effects of Cannabidiol, Hypothermia, and Their Combination in Newborn Rats with Hypoxic-Ischemic Encephalopathy. eNeuro 2023; 10:ENEURO.0417-22.2023. [PMID: 37072177 PMCID: PMC10166126 DOI: 10.1523/eneuro.0417-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/20/2023] Open
Abstract
Therapeutic hypothermia is well established as a standard treatment for infants with hypoxic-ischemic (HI) encephalopathy but it is only partially effective. The potential for combination treatments to augment hypothermic neuroprotection has major relevance. Our aim was to assess the effects of treating newborn rats following HI injury with cannabidiol (CBD) at 0.1 or 1 mg/kg, i.p., in normothermic (37.5°C) and hypothermic (32.0°C) conditions, from 7 d of age (neonatal phase) to 37 d of age (juvenile phase). Placebo or CBD was administered at 0.5, 24, and 48 h after HI injury. Two sensorimotor (rotarod and cylinder rearing) and two cognitive (novel object recognition and T-maze) tests were conducted 30 d after HI. The extent of brain damage was determined by magnetic resonance imaging, histologic evaluation, magnetic resonance spectroscopy, amplitude-integrated electroencephalography, and Western blotting. At 37 d, the HI insult produced impairments in all neurobehavioral scores (cognitive and sensorimotor tests), brain activity (electroencephalography), neuropathological score (temporoparietal cortexes and CA1 layer of hippocampus), lesion volume, magnetic resonance biomarkers of brain injury (metabolic dysfunction, excitotoxicity, neural damage, and mitochondrial impairment), oxidative stress, and inflammation (TNFα). We observed that CBD or hypothermia (to a lesser extent than CBD) alone improved cognitive and motor functions, as well as brain activity. When used together, CBD and hypothermia ameliorated brain excitotoxicity, oxidative stress, and inflammation, reduced brain infarct volume, lessened the extent of histologic damage, and demonstrated additivity in some parameters. Thus, coadministration of CBD and hypothermia could complement each other in their specific mechanisms to provide neuroprotection.
Collapse
Affiliation(s)
| | - Antonia A Alvarez
- Department of Cell Biology, University of the Basque Country, 48940 Leioa, Spain
| | - José J Rodríguez
- Functional Neuroanatomy Group, Biocruces Health Research Institute, 48903 Barakaldo, Spain
- Basque Foundation for Science (IKERBASQUE), 48009 Bilbao, Spain
- Department of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Hector Lafuente
- Biodonostia Health Research Institute, 20014 Donostia, Spain
| | - M Josune Canduela
- Department of Neurosciences, University of the Basque Country, 48940 Leioa, Spain
| | - William Hind
- Jazz Pharmaceuticals, Cambridge CB24 9BZ, United Kingdom
| | - José L Blanco-Bruned
- Department of Pediatric Surgery, Cruces University Hospital, OSI-Ezkerraldea Enkarterri Cruces, 48903 Barakaldo, Spain
| | | | - Enrique Hilario
- Department of Cell Biology, University of the Basque Country, 48940 Leioa, Spain
| |
Collapse
|
2
|
The utility of therapeutic hypothermia on cerebral autoregulation. JOURNAL OF INTENSIVE MEDICINE 2022; 3:27-37. [PMID: 36789361 PMCID: PMC9924009 DOI: 10.1016/j.jointm.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 11/07/2022]
Abstract
Cerebral autoregulation (CA) dysfunction is a strong predictor of clinical outcome in patients with acute brain injury (ABI). CA dysfunction is a potential pathologic defect that may lead to secondary injury and worse functional outcomes. Early therapeutic hypothermia (TH) in patients with ABI is controversial. Many factors, including patient selection, timing, treatment depth, duration, and rewarming strategy, impact its clinical efficacy. Therefore, optimizing the benefit of TH is an important issue. This paper reviews the state of current research on the impact of TH on CA function, which may provide the basis and direction for CA-oriented target temperature management.
Collapse
|
3
|
Ferreira RES, de Paiva BLC, de Freitas FGR, Machado FR, Silva GS, Raposo RM, Silveira CF, Centeno RS. Efficacy and Safety of a Nasopharyngeal Catheter for Selective Brain Cooling in Patients with Traumatic Brain Injury: A Prospective, Non-randomized Pilot Study. Neurocrit Care 2021; 34:581-592. [PMID: 32676873 DOI: 10.1007/s12028-020-01052-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/05/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The efficacy objective was to determine whether a novel nasopharyngeal catheter could be used to cool the human brain after traumatic brain injury, and the safety objective was to assess the local and systemic effects of this therapeutic strategy. METHODS This was a prospective, non-randomized, interventional clinical trial that involved five patients with severe traumatic brain injury. The intervention consisted of inducing and maintaining selective brain cooling for 24 h by positioning a catheter in the nasopharynx and circulating cold water inside the catheter in a closed-loop arrangement. Core temperature was maintained at ≥ 35 °C using counter-warming. RESULTS In all study participants, a brain temperature reduction of ≥ 2 °C was achieved. The mean brain temperature reduction from baseline was 2.5 ± 0.9 °C (P = .04, 95% confidence interval). The mean systemic temperature was 37.3 ± 1.1 °C at baseline and 36.0 ± 0.8 °C during the intervention. The mean difference between the brain temperature and the systemic temperature during intervention was - 1.2 ± 0.8 °C (P = .04). The intervention was well tolerated with no significant changes observed in the hemodynamic parameters. No relevant variations in intracranial pressure and transcranial Doppler were observed. The laboratory results underwent no major changes, aside from the K+ levels and blood counts. The K+ levels significantly varied (P = .04); however, the variation was within the normal range. Only one patient experienced an event of mild localized and superficial nasal discoloration, which was re-evaluated on the seventh day and indicated complete recovery. CONCLUSION The results suggest that our noninvasive method for selective brain cooling, using a novel nasopharyngeal catheter, was effective and safe for use in humans.
Collapse
Affiliation(s)
- Raphael Einsfeld Simões Ferreira
- Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, Av. Moema 170, Cj. 83. Moema, São Paulo, SP, 04077-020, Brazil.
| | | | | | - Flávia Ribeiro Machado
- Departamento de Anestesiologia, Dor e Terapia Intensiva, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gisele Sampaio Silva
- Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, Av. Moema 170, Cj. 83. Moema, São Paulo, SP, 04077-020, Brazil
| | - Rafael Mônaco Raposo
- Serviço de Otorrinolaringologia UNIFESP e Serviço de Otorrinolaringologia, Hospital Santa Paula, São Paulo, Brazil
| | - Conrado Feisthauer Silveira
- Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, Av. Moema 170, Cj. 83. Moema, São Paulo, SP, 04077-020, Brazil
| | - Ricardo Silva Centeno
- Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, Av. Moema 170, Cj. 83. Moema, São Paulo, SP, 04077-020, Brazil
| |
Collapse
|
4
|
Bay V, Happ DF, Ardalan M, Quist A, Oggiano F, Chumak T, Hansen K, Ding M, Mallard C, Tasker RA, Wegener G. Flinders sensitive line rats are resistant to infarction following transient occlusion of the middle cerebral artery. Brain Res 2020; 1737:146797. [PMID: 32194078 DOI: 10.1016/j.brainres.2020.146797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/17/2020] [Accepted: 03/14/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Depression is a common complication of stroke and increases the risk of mortality and disability. Pre-stroke depression is a possible risk factor for stroke and has also been linked to adverse outcomes. The underlying mechanisms linking depression and stroke remain unclear. Preclinical models may provide novel insights, but models reflecting both conditions are lacking. METHODS In this study, we investigated the effects of a 45-min transient middle cerebral artery occlusion (MCAo) on infarct size in male adult Flinders Sensitive Line rats, a genetic animal model of depression, and their control strains Flinders Resistant Line and Sprague-Dawley rats. Infarct size was assessed by tetrazolium chloride (TTC) and microtubule-associated protein 2 (MAP2) staining after 48 h of reperfusion. Angiograms of the vascular structure of naïve animals were produced with a µ-CT scanner. RESULTS Both Flinders strains had significantly smaller infarcts following MCAo compared to Sprague-Dawley rats. This effect does not appear to be due to changes in cerebrovascular architecture, as indicated by an initial exploration of vascular organization using angiograms, or body temperature regulation. CONCLUSIONS Our study suggests that the rat strain does not influence infarct volumes following MCAo.
Collapse
Affiliation(s)
- Vibeke Bay
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Denise F Happ
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark.
| | - Maryam Ardalan
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Physiology at Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Alexandra Quist
- Department of Physiology at Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Florian Oggiano
- Department of Physiology at Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Tetyana Chumak
- Department of Physiology at Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Kasper Hansen
- Comparative Medicine Lab, Department of Clinical Medicine, Health, Aarhus University, Denmark; Section for Zoophysiology, Department of Bioscience, Aarhus University, Denmark; Department of Forensic Medicine, Health, Aarhus University, Denmark
| | - Ming Ding
- Department of Orthopaedic Surgery & Traumatology, Odense University Hospital, Denmark; Department of Clinical Research, University of Southern Denmark
| | - Carina Mallard
- Department of Physiology at Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - R Andrew Tasker
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; Department of Biomedical Sciences, University of Prince Edward Island, Charlottetown PEI, Canada
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; Pharmaceutical Research Center of Excellence, School of Pharmacy (Pharmacology), North-West University, Potchefstroom, South Africa
| |
Collapse
|
5
|
Sun YJ, Zhang ZY, Fan B, Li GY. Neuroprotection by Therapeutic Hypothermia. Front Neurosci 2019; 13:586. [PMID: 31244597 PMCID: PMC6579927 DOI: 10.3389/fnins.2019.00586] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
Hypothermia therapy is an old and important method of neuroprotection. Until now, many neurological diseases such as stroke, traumatic brain injury, intracranial pressure elevation, subarachnoid hemorrhage, spinal cord injury, hepatic encephalopathy, and neonatal peripartum encephalopathy have proven to be suppressed by therapeutic hypothermia. Beneficial effects of therapeutic hypothermia have also been discovered, and progress has been made toward improving the benefits of therapeutic hypothermia further through combination with other neuroprotective treatments and by probing the mechanism of hypothermia neuroprotection. In this review, we compare different hypothermia induction methods and provide a summarized account of the synergistic effect of hypothermia therapy with other neuroprotective treatments, along with an overview of hypothermia neuroprotection mechanisms and cold/hypothermia-induced proteins.
Collapse
Affiliation(s)
- Ying-Jian Sun
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Zi-Yuan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Bin Fan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Guang-Yu Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Meuwly C, Chowdhury T, Sandu N, Golanov E, Erne P, Rosemann T, Schaller B. Definition and Diagnosis of the Trigeminocardiac Reflex: A Grounded Theory Approach for an Update. Front Neurol 2017; 8:533. [PMID: 29085328 PMCID: PMC5649131 DOI: 10.3389/fneur.2017.00533] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022] Open
Abstract
Background The trigeminocardiac reflex (TCR) is defined as sudden onset of parasympathetic dysrhythmias including hemodynamic irregularities, apnea, and gastric hypermotility during stimulation of sensory branches of the trigeminal nerve. Since the first description of the TCR in 1999, there is an ongoing discussion about a more emergent clinical definition. In this work, the author worked out an approach to such an improved definition. Methods In this study, a grounded theory approach was used. Literature about TCR was systematically identified through PubMed (MEDLINE), EMBASE (Ovid SP), and ISI Web of Sciences databases from 1/2005 until 8/2015. TCR was defined as a drop of heart rate (HR) below 60 bpm or 20% to the baseline. A grounded theory approach was used to analyze and interpret the data through a synthesis by the researcher’s perspectives, values, and positions. Results Out of the included studies, the authors formed available data to an update of the understanding of changes in hemodynamic parameters (HR and blood pressure) in a TCR. According to this update, an HR deceleration should be a constant observation to identify a TCR episode while a drop in blood pressure should probably not being fixed to a certain percentage of decrease. Conclusion The here presented working definition improves our understanding of the TCR. It leads the way to a new understanding of the TCR for a proper clinical definition.
Collapse
Affiliation(s)
| | - Tumul Chowdhury
- Department of Anaesthesiology and Perioperative Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Nora Sandu
- Department of Pathology, University of Buenos Aires, Buenos Aires, Argentina
| | - Eugene Golanov
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Paul Erne
- University Hospital Basel, Basel, Switzerland
| | - Thomas Rosemann
- Department of Primary Care, University of Zurich, Zürich, Switzerland
| | - Bernhard Schaller
- Department of Primary Care, University of Zurich, Zürich, Switzerland
| |
Collapse
|
7
|
Karakus Yilmaz B, Topcu H, Acar YA, Oran DS, Ozsoy S, Çevik E, Hakligor A, Çinar O. Optimum Temperature of Oxygen for Transpulmonary Hypothermia with Cooled Oxygen Inhalation: A Preliminary Study in a Rat Model. Ther Hypothermia Temp Manag 2016; 7:75-80. [PMID: 27849437 DOI: 10.1089/ther.2016.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cooled oxygen inhalation was hypothesized as a novel hypothermia technique in a previous study. In the current study, we aimed to determine the optimal temperature of oxygen for this method. This is a prospective, randomized, controlled, examiner-blinded experimental study conducted with 45 healthy, adult, Wistar Hannover male rats. Rats were randomly divided into five groups; group 1: +4°C intubated group (n = 7), group 2: +4°C nonintubated group (n = 9), group 3: +8°C intubated group (n = 9), group 4: +8°C nonintubated group (n = 9), and group 5: control group (n = 9). The control group received only a standardized anesthesia protocol, and no hypothermia technique was administered. Cooled oxygen was administered in the four study groups until the rectal temperature reached 34°C. The target temperature was maintained between 32°C and 34°C for 2 hours. Then, hypothermia protocols were terminated and rats were rewarmed externally with a blanket. Main outcomes were the speed (°C/minute) of temperature decrease (S) and the time required to reach the target body temperature (T). All study groups had better results than the control group in T and S values (p < 0.001) for both parameters. Group 1 had a better T value than group 4 (p = 0.01), but no difference in S value (p = 0.223). Comparison of group 2 and group 4 showed that group 4 had better results in T and S (p = 0.04 and 0.001, respectively). No pathologic changes in histologic examination were observed in any group. Our study showed that the optimal temperature of oxygen for the cooled oxygen technique was +4°C through an intubation tube.
Collapse
Affiliation(s)
- Banu Karakus Yilmaz
- 1 Department of Emergency Medicine, Hamidiye Sisli Etfal Training and Research Hospital , Istanbul, Turkey
| | - Hatice Topcu
- 1 Department of Emergency Medicine, Hamidiye Sisli Etfal Training and Research Hospital , Istanbul, Turkey
| | - Yahya A Acar
- 2 Department of Emergency Medicine, Etimesgut Military Hospital , Ankara, Turkey
| | - Duygu Sultan Oran
- 3 Experimental Research and Skills Development Center, Bagcilar Training and Research Hospital , Istanbul, Turkey
| | - Sule Ozsoy
- 4 Department of Pathology, Bagcilar Training and Research Hospital , Istanbul, Turkey
| | - Erdem Çevik
- 5 Department of Emergency Medicine, Van Military Hospital , Van, Turkey
| | - Aylin Hakligor
- 6 Department of Biochemistry, Adana Numune Training and Education Hospital , Adana, Turkey
| | - Orhan Çinar
- 7 Department of Emergency Medicine, Acıbadem University , Istanbul, Turkey
| |
Collapse
|
8
|
Ghahari L, Safari M, Joghataei MT, Mehdizadeh M, Soleimani M. Effect of combination therapy using hypothermia and granulocyte colony-stimulating factor in a rat transient middle cerebral artery occlusion model. IRANIAN BIOMEDICAL JOURNAL 2015; 18:239-44. [PMID: 25326023 PMCID: PMC4225064 DOI: 10.6091/ibj.13852.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Stroke is the third leading cause of death. Hypothermia has been recognized as an effective method in reducing brain injury. In this study, we assessed the effects of granulocyte colony-stimulating factor (G-CSF) as a neuroprotective agent and mild hypothermia on mortality, behavioral function, infarct volume, and brain edema in Wistar rats. Methods: Forty male rats were used in five groups (eight rats in each group): control, hypothermy, G-CSF, combination hypothermy + CSF, and sham. Rats were anesthetized by injection of chloral hydrate (400 mg/kg) intraperitoneally. Transient cerebral ischemia was induced by 60-min intraluminal occlusion of left middle cerebral artery. Hypothermia, initiated at the time of reperfusion and G-CSF was started one hour after reperfusion at a dose of 15 mg/kg subcutaneously. The motor behavior was measured using Garcia’s index and animals were assigned for the assessments of infarction, brain swelling, and mortality rate. Results: The mortality was 38.46% (control group) and reduced in other groups. Neurological deficit score of control group (40.31 ± 1.56) was significantly lower than in treatment groups. The total cerebral infarct volume of treatment group was significantly lower than control group (43.96 ± 44.05 mm3). Treatment with hypothermy plus G-CSF (2.69 ± 0.24%) could significantly reduce brain swelling volume than other treatment groups. Conclusion: Our major finding is that mild hypothermic treatment plus G-CSF significantly reduced mortality rate and edema and improved neurological function. The results suggest that the combination of hypothermia and G-CSF is more effectively than other treatment groups being used alone.
Collapse
Affiliation(s)
- Laya Ghahari
- Dept. of Anatomy, Medical School, Iran University of Medical Science, Tehran, Iran.,Dept. of Anatomy, Medical School, AJA University of Medical Sciences, Tehran, Iran
| | - Manouchehr Safari
- Dept. of Anatomy, Medical School, Semnan
University of Medical Science, Semnan, Iran
| | - Mohamad Taghi Joghataei
- Cellular and Molecular Research Center, Faculty of Advanced Technology in Medicine, Dept. of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Cellular and Molecular Research Center, Faculty of Advanced Technology in Medicine, Dept. of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Dept. of Anatomy, Medical School, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
9
|
Shah TA, Mauriello CT, Hair PS, Sandhu A, Stolz MP, Bass WT, Krishna NK, Cunnion KM. Clinical hypothermia temperatures increase complement activation and cell destruction via the classical pathway. J Transl Med 2014; 12:181. [PMID: 24962100 PMCID: PMC4079622 DOI: 10.1186/1479-5876-12-181] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 06/16/2014] [Indexed: 11/10/2022] Open
Abstract
Background Therapeutic hypothermia is a treatment modality that is increasingly used to improve clinical neurological outcomes for ischemia-reperfusion injury-mediated diseases. Antibody-initiated classical complement pathway activation has been shown to contribute to ischemia-reperfusion injury in multiple disease processes. However, how therapeutic hypothermia affects complement activation is unknown. Our goal was to measure the independent effect of temperature on complement activation, and more specifically, examine the relationship between clinical hypothermia temperatures (31–33°C), and complement activation. Methods Antibody-sensitized erythrocytes were used to assay complement activation at temperatures ranging from 0-41°C. Individual complement pathway components were assayed by ELISA, Western blot, and quantitative dot blot. Peptide Inhibitor of complement C1 (PIC1) was used to specifically inhibit activation of C1. Results Antibody-initiated complement activation resulting in eukaryotic cell lysis was increased by 2-fold at 31°C compared with 37°C. Antibody-initiated complement activation in human serum increased as temperature decreased from 37°C until dramatically decreasing at 13°C. Quantitation of individual complement components showed significantly increased activation of C4, C3, and C5 at clinical hypothermia temperatures. In contrast, C1s activation by heat-aggregated IgG decreased at therapeutic hypothermia temperatures consistent with decreased enzymatic activity at lower temperatures. However, C1q binding to antibody-coated erythrocytes increased at lower temperatures, suggesting that increased classical complement pathway activation is mediated by increased C1 binding at therapeutic hypothermia temperatures. PIC1 inhibited hypothermia-enhanced complement-mediated cell lysis at 31°C by up to 60% (P = 0.001) in a dose dependent manner. Conclusions In summary, therapeutic hypothermia temperatures increased antibody-initiated complement activation and eukaryotic cell destruction suggesting that the benefits of therapeutic hypothermia may be mediated via other mechanisms. Antibody-initiated complement activation has been shown to contribute to ischemia-reperfusion injury in several animal models, suggesting that for diseases with this mechanism hypothermia-enhanced complement activation may partially attenuate the benefits of therapeutic hypothermia.
Collapse
Affiliation(s)
- Tushar A Shah
- Department of Pediatrics, Eastern Virginia Medical School, 855 West Brambleton Avenue, Norfolk, VA 23510, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Chen F, Qi Z, Luo Y, Hinchliffe T, Ding G, Xia Y, Ji X. Non-pharmaceutical therapies for stroke: mechanisms and clinical implications. Prog Neurobiol 2014; 115:246-69. [PMID: 24407111 PMCID: PMC3969942 DOI: 10.1016/j.pneurobio.2013.12.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/19/2013] [Accepted: 12/27/2013] [Indexed: 12/14/2022]
Abstract
Stroke is deemed a worldwide leading cause of neurological disability and death, however, there is currently no promising pharmacotherapy for acute ischemic stroke aside from intravenous or intra-arterial thrombolysis. Yet because of the narrow therapeutic time window involved, thrombolytic application is very restricted in clinical settings. Accumulating data suggest that non-pharmaceutical therapies for stroke might provide new opportunities for stroke treatment. Here we review recent research progress in the mechanisms and clinical implications of non-pharmaceutical therapies, mainly including neuroprotective approaches such as hypothermia, ischemic/hypoxic conditioning, acupuncture, medical gases and transcranial laser therapy. In addition, we briefly summarize mechanical endovascular recanalization devices and recovery devices for the treatment of the chronic phase of stroke and discuss the relative merits of these devices.
Collapse
Affiliation(s)
- Fan Chen
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, Beijing 100053, China
| | - Zhifeng Qi
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, Beijing 100053, China
| | - Yuming Luo
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, Beijing 100053, China
| | - Taylor Hinchliffe
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - Guanghong Ding
- Shanghai Research Center for Acupuncture and Meridian, Shanghai 201203, China
| | - Ying Xia
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA.
| | - Xunming Ji
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, Beijing 100053, China.
| |
Collapse
|
12
|
Post cardiac arrest syndrome. COLOMBIAN JOURNAL OF ANESTHESIOLOGY 2014. [DOI: 10.1016/j.rcae.2014.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
13
|
Cornelius JF, Sandu N, Perez-Pinzon MA, Schaller B. Treatment of acute ischemic stroke: role of ischemic tolerance in intravenous and endovascular therapies. Expert Rev Cardiovasc Ther 2014; 7:331-2. [DOI: 10.1586/erc.09.18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
14
|
Post cardiac arrest syndrome☆. COLOMBIAN JOURNAL OF ANESTHESIOLOGY 2014. [DOI: 10.1097/01819236-201442020-00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
15
|
Coleman DM, Obi A, Criado E, Arya S, Berguer R. Contemporary outcomes after distal vertebral reconstruction. J Vasc Surg 2013; 58:152-7. [DOI: 10.1016/j.jvs.2012.12.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/27/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
|
16
|
Wozniak A, Mila-Kierzenkowska C, Szpinda M, Chwalbinska-Moneta J, Augustynska B, Jurecka A. Whole-body cryostimulation and oxidative stress in rowers: the preliminary results. Arch Med Sci 2013; 9:303-8. [PMID: 23671442 PMCID: PMC3648812 DOI: 10.5114/aoms.2012.30835] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 10/08/2011] [Accepted: 10/17/2011] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION The effect of whole-body cryostimulation (WBC) on the biomarkers of oxidative stress, lysosomal enzymes, creatine kinase and cortisol was studied. MATERIAL AND METHODS The rowers underwent two 6-day training cycles: with pre-training daily WBC (temperature: from -125°C to -150°C) and without cryostimulation (control). Blood samples were taken before and after the third and sixth day of training. RESULTS The activity of superoxide dismutase and glutathione peroxidase was lower (by 44% and 42%, respectively) after the third day of training with WBC than without WBC. The concentration of lipid peroxidation products was also lower after the training preceded by WBC. Moreover, the acid phosphatase activity was 50% lower after the third day of training with WBC than training without WBC. Considering the antioxidant enzymes activity during training without WBC, the increase of superoxide dismutase and glutathione peroxidase activity was observed after the third day of training (by about 74% and 100%, respectively). The level of lipid peroxidation products also increased after the training without WBC. No statistically significant changes were observed in creatine kinase activity after the training preceded with WBC, while after the training without WBC activity of this enzyme was two-fold higher than before the training. CONCLUSIONS The use of WBC prior to training may reduce the risk of oxidative stress and the extent of muscle fibre injuries provoked by intense exercise. The WBC seems to be an effective and safe method for limiting exercise-induced damage; thus it may be used in biological regeneration of sportsmen.
Collapse
Affiliation(s)
- Alina Wozniak
- The Chair of Medical Biology, Collegium Medicum of Nicolaus Copernicus University, Bydgoszcz, Poland
| | | | - Michał Szpinda
- Department of Normal Anatomy, Collegium Medicum of Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Jolanta Chwalbinska-Moneta
- Department of Applied Physiology, Medical Research Centre of the Polish Academy of Science, Warsaw, Poland
| | - Beata Augustynska
- Department of Psychiatry, Collegium Medicum of Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Alicja Jurecka
- The Chair of Medical Biology, Collegium Medicum of Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
17
|
Analysis of gene expression changes to elucidate the mechanism of chilling injury in precision-cut liver slices. Toxicol In Vitro 2012; 27:890-9. [PMID: 23111282 DOI: 10.1016/j.tiv.2012.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 10/10/2012] [Accepted: 10/11/2012] [Indexed: 11/22/2022]
Abstract
The exact mechanism of chilling injury (by a decrease of temperature to sub-physiological values), especially in the intact organ, is yet unknown. Precision-cut liver slices (PCLS), which closely resemble the organ from which they are derived, are an ideal in vitro model to study the mechanism of chilling injury in the intact organ. In the present study we were able to separate chilling injury from other damaging events such as cryoprotectant toxicity and ice-crystal injury and performed micro-array analysis of regulated genes. Pathway analysis revealed that different stress responses, lipid/fatty acid and cholesterol biosynthesis and metabolism were affected by chilling. This indicates that the cell-membrane might be the primary site and sensor for chilling, which may initiate and amplify downstream intracellular signaling events. Most importantly, we were able to identify gene expression responses from stellate cells and Kupffer cells suggesting the involvement of all liver cell types in the injury. In conclusion, a broad spectrum of previously unknown gene expression changes induced by chilling was identified in the tissue. This is the first report of a systematic investigation on the mechanism of chilling injury in integrated tissue by micro-array analysis under conditions in which other sources of injury are minimal.
Collapse
|
18
|
Zgavc T, Ceulemans AG, Hachimi-Idrissi S, Kooijman R, Sarre S, Michotte Y. The neuroprotective effect of post ischemic brief mild hypothermic treatment correlates with apoptosis, but not with gliosis in endothelin-1 treated rats. BMC Neurosci 2012; 13:105. [PMID: 22920191 PMCID: PMC3502503 DOI: 10.1186/1471-2202-13-105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 08/21/2012] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Stroke remains one of the most common diseases with a serious impact on quality of life but few effective treatments exist. Mild hypothermia (33°C) is a promising neuroprotective therapy in stroke management. This study investigated whether a delayed short mild hypothermic treatment is still beneficial as neuroprotective strategy in the endothelin-1 (Et-1) rat model for a transient focal cerebral ischemia. Two hours of mild hypothermia (33°C) was induced 20, 60 or 120 minutes after Et-1 infusion. During the experiment the cerebral blood flow (CBF) was measured via Laser Doppler Flowmetry in the striatum, which represents the core of the infarct. Functional outcome and infarct volume were assessed 24 hours after the insult. In this sub-acute phase following stroke induction, the effects of the hypothermic treatment on apoptosis, phagocytosis and astrogliosis were assessed as well. Apoptosis was determined using caspase-3 immunohistochemistry, phagocytic cells were visualized by CD-68 expression and astrogliosis was studied by glial fibrillary acidic protein (GFAP) staining. RESULTS Cooling could be postponed up to 1 hour after the onset of the insult without losing its positive effects on neurological deficit and infarct volume. These results correlated with the caspase-3 staining. In contrast, the increased CD-68 expression post-stroke was reduced in the core of the insult with all treatment protocols. Hypothermia also reduced the increased levels of GFAP staining, even when it was delayed up to 2 hours after the insult. The study confirmed that the induction of the hypothermia treatment in the Et-1 model does not affect the CBF. CONCLUSIONS These data indicate that in the Et-1 rat model, a short mild hypothermic treatment delayed for 1 hour is still neuroprotective and correlates with apoptosis. At the same time, hypothermia also establishes a lasting inhibitory effect on the activation of astrogliosis.
Collapse
Affiliation(s)
- Tine Zgavc
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neuroscience, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
19
|
Sadaka F, Veremakis C. Therapeutic hypothermia for the management of intracranial hypertension in severe traumatic brain injury: a systematic review. Brain Inj 2012; 26:899-908. [PMID: 22448655 DOI: 10.3109/02699052.2012.661120] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major source of death and severe disability worldwide. Raised Intracranial pressure (ICP) is an important predictor of mortality in patients with severe TBI and aggressive treatment of elevated ICP has been shown to reduce mortality and improve outcome. The acute post-injury period in TBI is characterized by several pathophysiologic processes that start in the minutes to hours following injury. All of these processes are temperature-dependent; they are all aggravated by fever and inhibited by hypothermia. METHODS This study reviewed the current clinical evidence in support of the use of therapeutic hypothermia (TH) for the treatment of intracranial hypertension (ICH) in patients with severe TBI. RESULTS This study identified a total of 18 studies involving hypothermia for control of ICP; 13 were randomized controlled trials (RCT) and five were observational studies. TH (32-34°C) was effective in controlling ICH in all studies. In the 13 RCT, ICP in the TH group was always significantly lower than ICP in the normothermia group. In the five observational studies, ICP during TH was always significantly lower than prior to inducing TH. CONCLUSIONS Pending results from large multi-centre studies evaluating the effect of TH on ICH and outcome, TH should be included as a therapeutic option to control ICP in patients with severe TBI.
Collapse
Affiliation(s)
- Farid Sadaka
- St. John's Mercy Medical Center, St Louis University, St Louis, MO, USA.
| | | |
Collapse
|
20
|
Allen BS, Ko Y, Buckberg GD, Tan Z. Studies of isolated global brain ischaemia: II. Controlled reperfusion provides complete neurologic recovery following 30 min of warm ischaemia - the importance of perfusion pressure. Eur J Cardiothorac Surg 2012; 41:1147-54. [PMID: 22436245 DOI: 10.1093/ejcts/ezr317] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES Neurologic injury after sudden death is likely due to a reperfusion injury following prolonged brain ischaemia, and remains problematic, especially if the cardiac arrest is unwitnessed. This study applies a newly developed isolated model of global brain ischaemia (simulating unwitnessed sudden death) for 30 min to determine if controlled reperfusion permits neurologic recovery. METHODS Among the 17 pigs undergoing 30 min of normothermic global brain ischaemia, 6 received uncontrolled reperfusion with regular blood (n = 6), and 11 were reperfused for 20 min with a warm controlled blood reperfusate containing hypocalcaemia, hyper-magnesemia, alkalosis, hyperosmolarty and other constituents that were passed through a white blood cell filter and delivered at flow rates of 350 cc/min (n = 3), 550 cc/min (n = 2) or 750 cc/min (n = 6). Neurologic deficit score (NDS) evaluated brain function (score 0 = normal, 500 = brain death) 24 h post-reperfusion and 2,3,5-triphenyltetrazolium chloride (TTC) staining determined brain infarction. RESULTS Regular blood (uncontrolled) reperfusion caused negligible brain O(2) uptake by IN Vivo Optical Spectroscopy (INVOS) (<10-15% O(2) extraction), oxidant damage demonstrated by raised conjugated diene (CD) levels (1.78 ± 0.13 A233 mn), multiple seizures, 1 early death from brain herniation, high NDS (249 ± 39) in survivors, brain oedema (84.4 ± 0.6%) and extensive cerebral infarctions. Conversely, controlled reperfusion restored surface brain oxygen saturation by INVOS to normal (55-70%), but the extent of neurologic recovery was determined by the brain reperfusion pressure. Low pressure reperfusion (independent of flow) produced the same adverse functional, metabolic and anatomic changes that followed uncontrolled reperfusion in seven pigs (three at 350 cc/min, two at 550 and two at 750 cc/min). Conversely, higher reperfusion pressure in four pigs (all at 750 cc/min) resulted in NDS of 0-70* indicating complete (n = 2) or near complete (n = 2) neurological recovery, negligible CDs production (1.29 ± 0.06 A233mn)*, minimal brain oedema (80.6 ± 0.2%)* and no infarction by TTC stain. CONCLUSIONS Brain injury can be avoided after 30 min of normothermic cerebral ischaemia if controlled reperfusion pressure is >50 mmHg, but the lower pressure (<50 mmHg) controlled reperfusion that is useful in other organs cannot be transferred to the brain. Moreover, INVOS is a poor guide to the adequacy of cerebral perfusion and the capacity of controlled brain reperfusion to restore neurological recovery. *P < 0.001 versus uncontrolled or low pressure controlled reperfusion.
Collapse
Affiliation(s)
- Bradley S Allen
- Department of Surgery, University of California, Los Angeles, CA, USA.
| | | | | | | |
Collapse
|
21
|
Allen BS, Buckberg GD. Studies of isolated global brain ischaemia: I. Overview of irreversible brain injury and evolution of a new concept - redefining the time of brain death. Eur J Cardiothorac Surg 2012; 41:1132-7. [PMID: 22398465 DOI: 10.1093/ejcts/ezr315] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite advanced cardiac life support (ACLS), the mortality from sudden death after cardiac arrest is 85-95%, and becomes nearly 100% if ischaemia is prolonged, as occurs following unwitnessed arrest. Moreover, 33-50% of survivors following ACLS after witnessed arrest develop significant neurological dysfunction, and this rises to nearly 100% in the rare survivors of unwitnessed arrest. Although, whole body (cardiac) survival improves to 30% following recent use of emergency cardiopulmonary bypass, sustained neurological dysfunction remains a devastating and unresolved problem. Our studies suggest that both brain and whole body damage reflect an ischaemic/reperfusion injury that follows the present reperfusion methods that use normal blood, which we term 'uncontrolled reperfusion'. In contrast, we have previously introduced the term 'controlled reperfusion', which denotes controlling both the conditions (pressure, flow and temperature) as well as the composition (solution) of the reperfusate. Following prolonged ischaemia of the heart, lung and lower extremity, controlled reperfusion resulted in tissue recovery after ischaemic intervals previously thought to produce irreversible cellular injury. These observations underlie the current hypothesis that controlled reperfusion will become an effective treatment of the otherwise lethal injury of prolonged brain ischaemia, such as with unwitnessed arrest, and we tested this after 30 min of normothermic global brain ischaemia. This review, and the subsequent three studies will describe the evolution of the concept that controlled reperfusion will restore neurological function to the brain following prolonged (30 min) ischaemia. To provide a familiarity and rationale for these studies, this overview reviews the background and current treatment of sudden death, the concepts of controlled reperfusion, recent studies in the brain during whole body ischaemia, and then summarizes the three papers in this series on a new brain ischaemia model that endorses our hypothesis that controlled reperfusion allows complete neurological recovery following 30 min of normothermic global brain ischaemia. These findings may introduce innovative management approaches for sudden death, and perhaps stroke, because the brain is completely salvageable following ischaemic times thought previously to produce infarction.
Collapse
Affiliation(s)
- Bradley S Allen
- Department of Surgery, University of California, Los Angeles, CA, USA.
| | | |
Collapse
|
22
|
Zgavc T, Ceulemans AG, Sarre S, Michotte Y, Hachimi-Idrissi S. Experimental and clinical use of therapeutic hypothermia for ischemic stroke: opportunities and limitations. Stroke Res Treat 2011; 2011:689290. [PMID: 21789271 PMCID: PMC3140058 DOI: 10.4061/2011/689290] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 05/11/2011] [Indexed: 01/04/2023] Open
Abstract
Stroke remains a disease with a serious impact on quality of life but few effective treatments exist. There is an urgent need to develop and/or improve neuroprotective strategies to combat this. Many drugs proven to be neuroprotective in experimental models fail to improve patient outcome in a clinical setting. An emerging treatment, therapeutic hypothermia (TH), is a promising neuroprotective therapy in stroke management. Several studies with TH in experimental models and small clinical trials have shown beneficial effects. Despite this, implementation into the clinical setting is still lacking due to methodological considerations as well as hypothermia-related complications. This paper discusses the possible opportunities and limitations of the use of TH in animal models and the translation into the clinic.
Collapse
Affiliation(s)
- Tine Zgavc
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neuroscience, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | | | | | | | | |
Collapse
|
23
|
Mitchell HM, White DM, Domowicz MS, Kraig RP. Cold pre-conditioning neuroprotection depends on TNF-α and is enhanced by blockade of interleukin-11. J Neurochem 2010; 117:187-96. [PMID: 21070241 DOI: 10.1111/j.1471-4159.2010.07103.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cold pre-conditioning reduces subsequent brain injury in small animals but the underlying mechanisms remain undefined. As hypothermia triggers systemic macrophage tumor necrosis factor alpha (TNF-α) production and other neural pre-conditioning stimuli depend on this cytokine, we reasoned that microglia and TNF-α would be similarly involved with cold pre-conditioning neuroprotection. Also, as slice cultures closely approximate their in vivo counterpart and include quiescent microglia, we used rat hippocampal slice cultures to confirm this hypothesis. Furthermore, inflammatory cytokine gene screening with subsequent PCR and immunostaining confirmation of targeted mRNA and related protein changes showed that cold pre-conditioning triggered a significant rise in TNF-α that localized to microglia and a significant rise in interleukin (IL)-11 that localized mainly to hippocampal pyramidal neurons and, more rarely, astrocytes. Importantly, co-stimulation with cold and IL-11, an anti-inflammatory cytokine that inhibits TNF-α expression, abrogated the otherwise evident protection. Instead, cold pre-conditioning coupled with blockade of IL-11 signaling further enhanced neuroprotection from that seen with cold pre-conditioning alone. Thus, physiological activation of brain pro-inflammatory cytokine signaling, and its amplification by inhibition of coincident anti-inflammatory cytokine signaling, may be opportune targets for the development of novel therapeutics that can mimic the protection seen in cold pre-conditioning.
Collapse
Affiliation(s)
- Heidi M Mitchell
- Department of Neurology, The University of Chicago Medical Center, Chicago, Illinois 60637, USA
| | | | | | | |
Collapse
|
24
|
Buckberg GD. Controlled reperfusion after ischemia may be the unifying recovery denominator. J Thorac Cardiovasc Surg 2010; 140:12-8, 18.e1-2. [DOI: 10.1016/j.jtcvs.2010.02.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 02/08/2010] [Indexed: 11/27/2022]
|
25
|
|
26
|
Aller MA, Arias JI, Alonso-Poza A, Arias J. A review of metabolic staging in severely injured patients. Scand J Trauma Resusc Emerg Med 2010; 18:27. [PMID: 20478066 PMCID: PMC2883961 DOI: 10.1186/1757-7241-18-27] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 05/17/2010] [Indexed: 02/07/2023] Open
Abstract
An interpretation of the metabolic response to injury in patients with severe accidental or surgical trauma is made. In the last century, various authors attributed a meaning to the post-traumatic inflammatory response by using teleological arguments. Their interpretations of this response, not only facilitates integrating the knowledge, but also the flow from the bench to the bedside, which is the main objective of modern translational research. The goal of the current review is to correlate the metabolic changes with the three phenotypes -ischemia-reperfusion, leukocytic and angiogenic- that the patients express during the evolution of the systemic inflammatory response. The sequence in the expression of multiple metabolic systems that becomes progressively more elaborate and complex in severe injured patients urges for more detailed knowledge in order to establish the most adequate metabolic support according to the evolutive phase. Thus, clinicians must employ different treatment strategies based on the different metabolic phases when caring for this challenging patient population. Perhaps, the best therapeutic option would be to favor early hypometabolism during the ischemia-reperfusion phase, to boost the antienzymatic metabolism and to reduce hypermetabolism during the leukocytic phase through the early administration of enteral nutrition and the modulation of the acute phase response. Lastly, the early epithelial regeneration of the injured organs and tissues by means of an oxidative metabolism would reduce the fibrotic sequelae in these severely injured patients.
Collapse
Affiliation(s)
- Maria-Angeles Aller
- Surgery I Department, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | | | | |
Collapse
|
27
|
Sandu N, Cornelius J, Filis A, Arasho B, Perez-Pinzon M, Schaller B. Ischemic tolerance in stroke treatment. Expert Rev Cardiovasc Ther 2010; 7:1255-61. [PMID: 19814668 DOI: 10.1586/erc.09.90] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although outcome after stroke treatment has significantly improved over the last 30 years, there has been no revolutionary breakthrough. Among different combined approaches, systemic thrombolysis in combination with neuroprotection became a favorite research target. Recent studies suggest that transient ischemic attacks may represent a clinical model of such ischemic tolerance; thus, a new focus on this research has emerged. In this review, we show the parallels between ischemia and neuroprotection and discuss the potential therapeutic options that may be opened by this new molecular knowledge.
Collapse
Affiliation(s)
- Nora Sandu
- Department of Neurosurgery, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
28
|
Batra RK, Paddle JJ. Therapeutic hypothermia in drowning induced hypoxic brain injury: a case report. CASES JOURNAL 2009; 2:9103. [PMID: 20062680 PMCID: PMC2803900 DOI: 10.1186/1757-1626-2-9103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 11/27/2009] [Indexed: 11/30/2022]
Abstract
Background Although therapeutic hypothermia for neuroprotection has been in use for over half a century but its use has been controversial in absence of proper guidelines. However for over two decades there has been revived interest in mild therapeutic hypothermia (32 - 34°C) for neuroprotection. Case A 17 year-old female tourist was rescued from sea. She received cardio-pulmonary resuscitation for about 16 minutes. But she had sustained significant neurological insult as a result of hypoxic brain injury. Therapeutic hypothermia was added to her regime of neuroprotection in intensive care unit, and her neurological status improved in just 8 hours with full correction of her coma score by day 4.
Collapse
Affiliation(s)
- Ramesh K Batra
- Intensive Care Unit, Royal Cornwall Hospital NHS Trust, Truro, UK
| | | |
Collapse
|
29
|
Abstract
BACKGROUND Mild to moderate hypothermia (32-35 degrees C) is the first treatment with proven efficacy for postischemic neurological injury. In recent years important insights have been gained into the mechanisms underlying hypothermia's protective effects; in addition, physiological and pathophysiological changes associated with cooling have become better understood. OBJECTIVE To discuss hypothermia's mechanisms of action, to review (patho)physiological changes associated with cooling, and to discuss potential side effects. DESIGN Review article. INTERVENTIONS None. MAIN RESULTS A myriad of destructive processes unfold in injured tissue following ischemia-reperfusion. These include excitotoxicty, neuroinflammation, apoptosis, free radical production, seizure activity, blood-brain barrier disruption, blood vessel leakage, cerebral thermopooling, and numerous others. The severity of this destructive cascade determines whether injured cells will survive or die. Hypothermia can inhibit or mitigate all of these mechanisms, while stimulating protective systems such as early gene activation. Hypothermia is also effective in mitigating intracranial hypertension and reducing brain edema. Side effects include immunosuppression with increased infection risk, cold diuresis and hypovolemia, electrolyte disorders, insulin resistance, impaired drug clearance, and mild coagulopathy. Targeted interventions are required to effectively manage these side effects. Hypothermia does not decrease myocardial contractility or induce hypotension if hypovolemia is corrected, and preliminary evidence suggests that it can be safely used in patients with cardiac shock. Cardiac output will decrease due to hypothermia-induced bradycardia, but given that metabolic rate also decreases the balance between supply and demand, is usually maintained or improved. In contrast to deep hypothermia (<or=30 degrees C), moderate hypothermia does not induce arrhythmias; indeed, the evidence suggests that arrhythmias can be prevented and/or more easily treated under hypothermic conditions. CONCLUSIONS Therapeutic hypothermia is a highly promising treatment, but the potential side effects need to be properly managed particularly if prolonged treatment periods are required. Understanding the underlying mechanisms, awareness of physiological changes associated with cooling, and prevention of potential side effects are all key factors for its effective clinical usage.
Collapse
|
30
|
Therapeutic hypothermia and controlled normothermia in the intensive care unit: practical considerations, side effects, and cooling methods. Crit Care Med 2009; 37:1101-20. [PMID: 19237924 DOI: 10.1097/ccm.0b013e3181962ad5] [Citation(s) in RCA: 476] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hypothermia is being used with increasing frequency to prevent or mitigate various types of neurologic injury. In addition, symptomatic fever control is becoming an increasingly accepted goal of therapy in patients with neurocritical illness. However, effectively controlling fever and inducing hypothermia poses special challenges to the intensive care unit team and others involved in the care of critically ill patients. OBJECTIVE To discuss practical aspects and pitfalls of therapeutic temperature management in critically ill patients, and to review the currently available cooling methods. DESIGN Review article. INTERVENTIONS None. MAIN RESULTS Cooling can be divided into three distinct phases: induction, maintenance, and rewarming. Each has its own risks and management problems. A number of cooling devices that have reached the market in recent years enable reliable maintenance and slow and controlled rewarming. In the induction phase, rapid cooling rates can be achieved by combining cold fluid infusion (1500-3000 mL 4 degrees C saline or Ringer's lactate) with an invasive or surface cooling device. Rapid induction decreases the risks and consequences of short-term side effects, such as shivering and metabolic disorders. Cardiovascular effects include bradycardia and a rise in blood pressure. Hypothermia's effect on myocardial contractility is variable (depending on heart rate and filling pressure); in most patients myocardial contractility will increase, although mild diastolic dysfunction can develop in some patients. A risk of clinically significant arrhythmias occurs only if core temperature decreases below 30 degrees C. The most important long-term side effects of hypothermia are infections (usually of the respiratory tract or wounds) and bedsores. CONCLUSIONS Temperature management and hypothermia induction are gaining importance in critical care medicine. Intensive care unit physicians, critical care nurses, and others (emergency physicians, neurologists, and cardiologists) should be familiar with the physiologic effects, current indications, techniques, complications and practical issues of temperature management, and induced hypothermia. In experienced hands the technique is safe and highly effective.
Collapse
|
31
|
Schaller B. Extracranial-intracranial bypass to reduce the risk of ischemic stroke in intracranial aneurysms of the anterior cerebral circulation: a systematic review. J Stroke Cerebrovasc Dis 2009; 17:287-98. [PMID: 18755409 DOI: 10.1016/j.jstrokecerebrovasdis.2008.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Revised: 03/17/2008] [Accepted: 03/27/2008] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE If clip application or coil placement for treatment of intracranial (IC) aneurysms is not feasible, the parent vessel can be occluded to induce thrombosis of the aneurysm. In the case that such an occlusion cannot be tolerated without subsequent sequel, the additional construction of an extracranial (EC)-IC bypass is needed for sufficient ipsilateral revascularization. Hitherto, the effectiveness of this combined treatment option was not investigated in a controlled randomized trial or in a review. The aim of the current report was to analyze clinical effectiveness of EC-IC bypass for cerebral revascularization in patients with Hunterian ligation in case of otherwise untreatable aneurysm of the anterior cerebral circulation. Special reference was given to different hemodynamic subgroups. METHODS A computerized database search was conducted from November 1985 to November 2002 using MEDLINE, relevant Internet sources, and full-text journal articles using appropriate indexed terms. Journal of Neurosurgery, Neurosurgery, Acta Neurochirurgica, and Stroke were manually searched for the period November 1985 to November 2002 and checked reference lists of all relevant articles for additional eligible studies. Language restriction was done for English, French, and German. Reports dealing with EC-IC bypass surgery for cerebral revascularization in case of aneurysm of the anterior cerebral circulation were reviewed when appropriate. Studies were included that contained evaluable data on clinical state, preoperative and postoperative hemodynamic state, surgical outcome, and follow-up. A statistical analysis was performed for different outcome parameters and clinical effectiveness in the included studies. RESULTS Overall, 20 studies were included, each with a study quality of 0-1. The postoperative outcome related to death or stroke depended mainly on preoperative hemodynamic subgroups (cerebral blood flow [CBF]/cerebral blood volume [CBV]; oxygen extraction fraction [OEF]). The final functional status was worse the more CBF/CBV ratio and OEF increased. Perioperative risk for death (0.8%) or stroke (1.5%) during the first month after operation was similar to the death or stroke rate during the following 2 to 12 months after operation. Neurologic function was improved over the preoperative state in 74% of the patients and was unchanged in 9%. The modified Rankin scale score was postoperatively 0 to 1 in 81% and 2 in 6% of the patients. Long-term patency was excellent, with 2.3% failure rate per year after the first year after surgery. There was no de novo aneurysm formation in the follow-up. CONCLUSION Neurologic function and subsequent stroke attributable to hemodynamic insufficiency in patients with otherwise untreatable IC aneurysm improves significantly by EC-IC bypass surgery if the brain area corresponding to the impaired neurologic function remains viable. The hemodynamic parameters observed for patients who experience improved neurologic function or diminished stroke risk profile after EC-IC bypass surgery contain both significantly elevated OEF and CBF/CBV. Therefore, hemodynamic state represents an important indicator for EC-IC bypass surgery. The large amount of data leads to narrow stroke with no significant heterogeneity, and the overall results are, therefore, likely to be statistically robust.
Collapse
|
32
|
Nagel S, Papadakis M, Hoyte L, Buchan AM. Therapeutic hypothermia in experimental models of focal and global cerebral ischemia and intracerebral hemorrhage. Expert Rev Neurother 2008; 8:1255-68. [PMID: 18671669 DOI: 10.1586/14737175.8.8.1255] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Experimental evidence shows that therapeutic hypothermia (TH) protects the brain from cerebral injury in multiple ways. In different models of focal and global cerebral ischemia, mild-to-moderate hypothermia reduces mortality and neuronal injury and improves neurological outcome. In models of experimental intracerebral hemorrhage (ICH), TH reduces edema formation but does not show consistent benefi cial effects on functional outcome parameters. However, the number of studies of hypothermia on ICH is still limited. TH is most effective when applied before or during the ischemic event, and its neuroprotective properties vary according to species, strains and the model of ischemia used. Intrinsic changes in body and brain temperature frequently occur in experimental models of focal and global cerebral ischemia, and may have infl uenced studies on other neuroprotectants. This might be one explanation for the failure of a large amount of translational clinical neuroprotective trials. Hypothermia is the only neuroprotective therapeutic agent for cerebral ischemia that has successfully managed the transfer from bench to bedside, and it is an approved therapy for patients after cardiac arrest and children with hypoxic-ischemic encephalopathy. However, the implementation of hypothermia in the treatment of stroke patients is still far from routine clinical practice. In this article, the authors describe the development of TH in different models of focal and global cerebral ischemia, point out why hypothermia is so efficient in experimental cerebral ischemia, explain why temperature regulation is essential for further neuroprotective studies and discuss why TH for acute ischemic stroke still remains a promising but controversial therapeutic option.
Collapse
Affiliation(s)
- Simon Nagel
- Acute Stroke Programme, Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | | | | | | |
Collapse
|
33
|
Lasater M. Intravascular temperature modulation as an adjunct to secondary brain injury prevention in a patient with an epidural hematoma. J Neurosci Nurs 2008; 40:198-200. [PMID: 18727335 DOI: 10.1097/01376517-200808000-00003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Epidural hematomas (EDHs) account for 10% of fatal injuries in patients with head trauma. Up to 35% of patients suffering EDHs develop secondary brain injury within days after the initial trauma. Secondary brain injury can profoundly affect a patient's subsequent neurologic recovery and functional status. This case study presents a patient who developed an EDH following severe head trauma with signs and symptoms of secondary brain injury; intravascular temperature modulation was used to treat the secondary injury for a prolonged period of 13 days. The patient showed a complete neurologic recovery with return to work 6 months after her injury.
Collapse
|
34
|
Wong AA, Read SJ. Early changes in physiological variables after stroke. Ann Indian Acad Neurol 2008; 11:207-20. [PMID: 19893676 PMCID: PMC2771993 DOI: 10.4103/0972-2327.44555] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 09/23/2008] [Accepted: 09/23/2008] [Indexed: 01/04/2023] Open
Abstract
Several aspects of physiology, notably blood pressure, body temperature, blood glucose, and blood oxygen saturation, may be altered after an ischemic stroke and intracerebral hemorrhage. Generally, blood pressure and temperature rise acutely after a stroke, before returning to normal. Blood glucose and oxygen levels may be abnormal in individuals, but they do not follow a set pattern. Several aspects of these physiological alterations remain unclear, including their principal determinants - whether they genuinely affect prognosis (as opposed to merely representing underlying processes such as inflammation or a stress response), whether these effects are adaptive or maladaptive, whether the effects are specific to certain subgroups (e.g. lacunar stroke) and whether modifying physiology also modifies its prognostic effect. Hypertension and hyperglycemia may be helpful or harmful, depending on the perfusion status after an ischemic stroke; the therapeutic response to their lowering may be correspondingly variable. Hypothermia may provide benefits, in addition to preventing harm through protection from hyperthermia. Hypoxia is harmful, but normobaric hyperoxia is unhelpful or even harmful in normoxic patients. Hyperbaric hyperoxia, however, may be beneficial, though this remains unproven. The above-mentioned uncertainties necessitate generally conservative measures for physiology management, although there are notably specific recommendations for thrombolysis-eligible patients. Stroke unit care is associated with better outcome, possibly through better management of poststroke physiology. Stroke units can also facilitate research to clarify the relationship between physiology and prognosis, and to subsequently clarify management guidelines.
Collapse
Affiliation(s)
- Andrew A Wong
- Department of Neurology, Princess Alexandra Hospital, Brisbane, Queensland, Australia. Central Clinical School, School of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| | | |
Collapse
|
35
|
Campbell K, Meloni BP, Knuckey NW. Combined magnesium and mild hypothermia (35 degrees C) treatment reduces infarct volumes after permanent middle cerebral artery occlusion in the rat at 2 and 4, but not 6 h. Brain Res 2008; 1230:258-64. [PMID: 18644354 DOI: 10.1016/j.brainres.2008.06.110] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 06/25/2008] [Accepted: 06/27/2008] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE Using transient focal and global cerebral ischemia models in the rat, we have previously shown that MgSO4 is not neuroprotective unless it is combined with mild hypothermia. This study establishes a therapeutic time window for combined MgSO4 and mild hypothermia treatment after permanent middle cerebral artery occlusion (MCAO). METHODS Rats were subjected to permanent intraluminal thread MCAO and animals were treated 2, 4 or 6 h after ischemia with a MgSO4 infusion (360 micromol/kg, then 120 micromol/kg/h) and mild hypothermia (35 degrees C) or with vehicle for 24 h. At the 2 h time point, treatment with hypothermia alone and MgSO4 alone were also assessed. Infarct volumes were measured 48 h after MCAO induction. RESULTS After permanent MCAO, combined MgSO4 and hypothermia treatment reduced infarct volumes by 54% at 2 h (P = 0.048) and by 39% at 4 h (P = 0.012), but there was no treatment effect detected at 6 h or in the hypothermia alone or MgSO4 alone groups. CONCLUSIONS These findings support our earlier work highlighting the neuroprotective effect of MgSO4 when combined with mild hypothermia, even when treatment is delayed by several hours.
Collapse
Affiliation(s)
- Kym Campbell
- Centre for Neuromuscular and Neurological Disorders/University of Western Australia, Australian Neuromuscular Research Institute, Department of Neurosurgery/Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | | | | |
Collapse
|
36
|
Grillon E, Provent P, Montigon O, Segebarth C, Rémy C, Barbier EL. Blood-brain barrier permeability to manganese and to Gd-DOTA in a rat model of transient cerebral ischaemia. NMR IN BIOMEDICINE 2008; 21:427-436. [PMID: 17948222 DOI: 10.1002/nbm.1206] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Loss of integrity of the blood-brain barrier (BBB) and brain swelling is a potentially lethal complication of reperfusion in human stroke. To assess the time course of BBB modifications, we performed angiography, diffusion-weighted imaging, T1-weighted (T1 W) imaging and T1 mapping, and monitored acute changes after middle cerebral artery occlusion and recanalization in rats (n = 27). The animals were grouped according to the duration of occlusion: 30 min (group A, n = 8), 1 h 30 min (group B, n = 9), and 2 h 30 min (group C, n = 10). For 17 animals (four in group A, six in group B, and seven in group C), MnCl2 and dimeglumine gadoterate (Gd-DOTA) were injected at 13 min and 34 min after recanalization, respectively. The 10 remaining animals (control groups) underwent the same acquisition protocols, but no contrast agents were injected. Cell damage was determined 1 h after recanalization on haematoxylin and eosin-stained sections. Our results indicate that in the middle cerebral artery occlusion model in the rat, changes in BBB permeability assessed by contrast agent extravasation occur within the first hour of reperfusion, even after an occlusion period not exceeding 30 min. No differences between BBB permeability to Gd-DOTA and Mn2+ were detected in our experimental conditions. The reduction in apparent diffusion coefficient during occlusion appears to be a good predictor of BBB modifications after reperfusion in this model.
Collapse
|
37
|
Nakano H, Colli BO, Lopes LDS. Neuroprotective effect of mild hypothermia in the temporary brain ischemia in cats. ARQUIVOS DE NEURO-PSIQUIATRIA 2008; 65:810-5. [PMID: 17952286 DOI: 10.1590/s0004-282x2007000500015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Accepted: 06/21/2007] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate the neuroprotective effect of mild hypothermia during temporary focal ischemia in cats. METHOD 20 cats underwent middle cerebral artery 60 minutes occlusion and 24 hours reperfusion: 10 under normothermia and 10 under mild hypothermia (32 masculine C). Brain coronal sections 2mm thick were stained with 2,3,5-triphenyltetrazolium hydrochloride, photographed and evaluated with software for volume calculation. RESULTS Cortical ischemia was found in 7 and basal ganglia ischemia in 8 animals of group 1 and in both regions in 5 animals of group 2 (no difference: p=0.6499 for cortical; p=0.3498 for basal ganglia). No ischemia was found in 5 animals of group 2 and in none of group 1 (significant difference, p=0.0325). The infarct volume was greater in group 1 than 2 (p=0.0433). CONCLUSION Mild hypothermia did not interfere with location of ischemia, but it was effective for reducing the infarct volume.
Collapse
Affiliation(s)
- Hiroshi Nakano
- Division of Neurosurgery, Department of Surgery and Anatomy, Hospital das Clinicas, Ribeirão Preto Medical School, University of São Paulo, 14048-900 Ribeirão Preto, SP, Brazil
| | | | | |
Collapse
|
38
|
Polderman KH. Induced Hypothermia for Neuroprotection: Understanding the Underlying Mechanisms. Intensive Care Med 2007. [DOI: 10.1007/0-387-35096-9_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
39
|
Ly JV, Arakawa S, Ma H, Donnan GA. Future advances in the treatment of acute ischemic stroke. FUTURE NEUROLOGY 2007. [DOI: 10.2217/14796708.2.1.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The main obstacles to the use of tissue plasminogen activator in acute ischemic stroke are its narrow therapeutic window of 3 h and the possibility of symptomatic intracranial hemorrhage. Hence, current research is directed towards extending the therapeutic window whilst minimizing this complication. This review summarizes different approaches and gives a brief overview of the current concepts that are important in order to overcome these limitations. The main issues discussed include the concept of ischemic penumbra and its salvage by means of improving patient selection based on penumbral imaging, reperfusion and neuroprotection. We provide a perspective into how current acute stroke research may shape future acute stroke therapy.
Collapse
Affiliation(s)
- John V Ly
- National Stroke Research Institute, Level 1, Neurosciences Building, Austin Health, University of Melbourne, 300 Waterdale Rd, Heidleberg Heights, Victoria 3081, Australia
| | - Shuji Arakawa
- National Stroke Research Institute, Level 1, Neurosciences Building, Austin Health, University of Melbourne, 300 Waterdale Rd, Heidleberg Heights, Victoria 3081, Australia
| | - Henry Ma
- National Stroke Research Institute, Level 1, Neurosciences Building, Austin Health, University of Melbourne, 300 Waterdale Rd, Heidleberg Heights, Victoria 3081, Australia
| | - Geoffrey A Donnan
- National Stroke Research Institute, Level 1, Neurosciences Building, Austin Health, University of Melbourne, 300 Waterdale Rd, Heidleberg Heights, Victoria 3081, Australia
| |
Collapse
|
40
|
Abstract
A hypothesis about the inflammatory etiopathogeny mediated by astroglia of hepatic encephalopathy is being proposed. Three evolutive phases are considered in chronic hepatic encephalopathy: an immediate or nervous phase with ischemia-reperfusion, which is associated with reperfusion injury, edema and oxidative stress; an intermediate or immune phase with microglia hyperactivity, which produces cytotoxic cytokines and chemokines and is involved in enzyme hyperproduction and phagocytosis; and a late or endocrine phase, in which neuroglial remodeling, with an alteration of angiogenesis and neurogenesis, stands out. The increasingly complex trophic meaning that the metabolic alterations have in the successive phases making up this chronic inflammation could explain the metabolic regression produced in acute and acute-on-chronic hepatic encephalopathy. In these two types of hepatic encephalopathy, characterized by edema, neuronal nutrition by diffusion would guarantee an appropriate support of substrates, in accordance with the reduced metabolic needs of the cerebral tissue.
Collapse
Affiliation(s)
- Jorge-Luis Arias
- Psychobiology Laboratory, School of Psychology, University of Oviedo, Asturias, Spain
| | | | | | | |
Collapse
|
41
|
Abstract
OBJECTIVE Dysphagia is common after stroke and represents a marker of poor prognosis. After ischemic stroke, dysphagia represents only one part of the clinical spectrum of changes in the gastrointestinal (GI) tract and includes GI hemorrhage, delayed GI emptying, and colorectal dysfunction. State-of-the-art imaging techniques have started to revolutionize to study the cortical and brainstem control of these GI symptoms. It has become increasingly obvious that GI alterations after stroke are complex and its recovery following stroke is even more so. METHODS In this review, an electronic database research was performed in MEDLINE, EMBASE, and the COCHRANE database using the terms stroke, dysphagia, GI motility, or cortical reorganization; an extensive manual searching was additionally conducted. RESULTS Cerebral ischemia may lead to an interruption of the axis between central nervous system and GI system. This altered interrelation between the central nervous system and the GI system may cause, among other things, mainly dysphagia, GI dysmotility, and GI hemorrhage. The consecutive clinical symptoms can often be directly attributed to specific cerebral ischemic lesions involving the brain stem as well as certain cortical and subcortical structures. However, in some cases the pathophysiological mechanisms leading to GI symptoms are incompletely understood. Recent improvement of imaging techniques, especially in functional imaging, has lead to new insights of the central control of the GI tract, suggesting that its cortical and medullar organization is multifocal, and bilateral with handness-independent hemispheric dominance. CONCLUSIONS Following stroke, patients may have swallowing impairment and other changes of the GI tract that could affect nutritional and hydration status and that lead to aspiration pneumonia. Impaired nutritional status is associated with reduced functional improvement, increased complication rates, and prolonged hospital stays.
Collapse
|
42
|
Abstract
Primary brain tumors are generally difficult to treat because of the unique location of the lesions. In addition, normal brain structures are often destroyed by the growing neoplasm. Even with effective therapy to surgically resect and destroy the neoplastic tissues, the brain is sometimes still injured, which can leave the patient in a debilitated state. The hemodynamic and metabolic state of such peritumoral brain tissue is not yet well understood, and there are only a small number of experimental hypotheses of its reaction and changes to the growing primary brain tumor. In addition, primary brain tumors may be influenced by certain anticancer drugs, which cause oxidative stress and consecutive cell death, or by gamma-irradiation. Currently, no established diagnostic methods exist to demonstrate and/or quantify the metabolic condition of the peritumoral tissue. The therapeutic strategy for possible pharmacological neuroprotection should, in the future, still be related to metabolic parameters, as well as in the peritumor tissue to treat primary brain tumors without risk to sensitive normal tissue. To achieve this aim, there has been particular emphasis on the biological behavior of primary brain tumors and peritumor tissue, as well as the potential correlation among them. Thus, priority should be given to identifying more target antigens in primary brain tumors and defining those cells present in the brain parenchyma that are essential to maintain a neuroprotective effect. However, at this time, the postinjury enhancement of neurogenesis appears to offer the best hope for long-lasting functional recovery following surgery of primary brain tumors.
Collapse
|
43
|
Duszczyk M, Gadamski R, Ziembowicz A, Lazarewicz JW. Antagonists of group I metabotropic glutamate receptors do not inhibit induction of ischemic tolerance in gerbil hippocampus. Neurochem Int 2006; 48:478-84. [PMID: 16524644 DOI: 10.1016/j.neuint.2005.12.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Revised: 12/19/2005] [Accepted: 12/22/2005] [Indexed: 10/24/2022]
Abstract
In this study we tested the effect of antagonists of two subtypes of the group I metabotropic glutamate receptors (mGluRs GI) on the induction of ischemic tolerance in relation to brain temperature. These experiments were prompted by indications that glutamate receptors may participate in the mechanisms of ischemic preconditioning. The role of NMDA receptors in the induction of ischemic tolerance has been debated while there is lack of information concerning the involvement of mGluRs GI in this phenomenon. The tolerance to injurious 3 min forebrain ischemia in Mongolian gerbils was induced 48 h earlier by 2 min preconditioning ischemia. Brain temperature was measured using telemetry equipment. EMQMCM and MTEP, antagonists of mGluR1 and mGluR5, respectively, were injected i.p. at a dose of 5 mg/kg. They were administered either before preconditioning ischemia in a single dose or after 2 min ischemia three times every 2 h. Both antagonists did not inhibit the induction of ischemic tolerance. Thus, our data indicate that group I metabotropic glutamate receptors do not play an essential role in the induction of ischemic tolerance.
Collapse
Affiliation(s)
- Malgorzata Duszczyk
- Department of Neurochemistry, Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
44
|
Schaller B, Graf R. Different compartments of intracranial pressure and its relationship to cerebral blood flow. ACTA ACUST UNITED AC 2006; 59:1521-31. [PMID: 16394936 DOI: 10.1097/01.ta.0000197402.20180.6b] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The classical "Kellie-Monroe" doctrine considering the intracranial volume to be a closed system that is confined within the nearly rigid skull, conserves different mass, and has equal vascular inflow and outflow. Several experimental and clinical studies have given evidence that this is not entirely true from the (patho)physiologic point of view, even so our understanding of this phenomenon is incomplete. METHODS Review from the literature. RESULTS The present literature review revokes this classical doctrine and suggests a more differentiated description for the dynamic of intracranial pressure (ICP): instead of the previously suggested lumped-parameter models, the authors describe different intracranial compartments that are related to different brain regions. CONCLUSION This has the advantage of great practical use on the one hand and allows the demonstration of relevant intercompartimental intracranial pressure differences. In addition, these ICP differences can be revealed to different ICP compartments and to its relationship to CBF. Special reference is given to determine appropriate forms for the nonconstant resistance and compliance parameters.
Collapse
Affiliation(s)
- B Schaller
- Max Planck Institute for Neurological Research, Cologne, Germany
| | | |
Collapse
|
45
|
|
46
|
van Breda EJ, van der Worp HB, van Gemert HMA, Algra A, Kappelle LJ, van Gijn J, Koudstaal PJ, Dippel DWJ. PAIS: paracetamol (acetaminophen) in stroke; protocol for a randomized, double blind clinical trial [ISCRTN 74418480]. BMC Cardiovasc Disord 2005; 5:24. [PMID: 16109181 PMCID: PMC1208871 DOI: 10.1186/1471-2261-5-24] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Accepted: 08/19/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In patients with acute stroke, increased body temperature is associated with large lesion volumes, high case fatality, and poor functional outcome. A 1 degrees C increase in body temperature may double the odds of poor outcome. Two randomized double-blind clinical trials in patients with acute ischemic stroke have shown that treatment with a daily dose of 6 g acetaminophen (paracetamol) results in a small but rapid and potentially worthwhile reduction of 0.3 degrees C (95% CI: 0.1-0.5) in body temperature. We set out to test the hypothesis that early antipyretic therapy reduces the risk of death or dependency in patients with acute stroke, even if they are normothermic. METHODS/DESIGN Paracetamol (Acetaminophen) In Stroke (PAIS) is a randomized, double-blind clinical trial, comparing high-dose acetaminophen with placebo in 2500 patients. Inclusion criteria are a clinical diagnosis of hemorrhagic or ischemic stroke and the possibility to start treatment within 12 hours from onset of symptoms. The study will have a power of 86% to detect an absolute difference of 6% in the risk of death or dependency at three months, and a power of 72% to detect an absolute difference of 5%, at a 5% significance level. DISCUSSION This is a simple trial, with a drug that only has a small effect on body temperature in normothermic patients. However, when lowering body temperature with acetaminophen does have the expected effectiveness, 20 patients will have to be treated to prevent dependency or death in one.
Collapse
Affiliation(s)
| | | | | | - Ale Algra
- University Medical Center Utrecht, the Netherlands
| | | | - Jan van Gijn
- University Medical Center Utrecht, the Netherlands
| | | | | | | |
Collapse
|
47
|
Alexandrova ML, Bochev PG. Oxidative stress during the chronic phase after stroke. Free Radic Biol Med 2005; 39:297-316. [PMID: 15993329 DOI: 10.1016/j.freeradbiomed.2005.04.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 04/15/2005] [Accepted: 04/20/2005] [Indexed: 12/20/2022]
Abstract
Stroke is a complex disease originating and developing on the background of genetic predisposition and interaction between different risk factors that chronically damage blood vessels. The search for an effective treatment of stroke patients is the main priority of basic and clinical sciences. The chronic phase of stroke provides possibilities for therapy directed toward stimulation of recovery processes as well as prophylaxis, which reduces the probability of subsequent cerebrovascular events. Oxidative stress is a potential contributor to the pathophysiological consequences of stroke. The aim of the present review is to summarize the current knowledge of the role of oxidative stress during the chronic phase after stroke and its contribution to the initiation of subsequent stroke. The relationship among inflammation, hemostatic abnormalities, and platelet activation in chronic stroke patients is discussed in the context of ongoing free radical processes and oxidative damage. Free radical-mediated effects of increased plasma level of homocysteine and its possible contribution to the processes leading to recurrent stroke are discussed as well. The status of the antioxidant defense system and the degree of oxidative damage in the circulation of stroke survivors are examined. The results are interpreted in view of the effects of the vascular risk factors for stroke that include additional activation of inflammatory and free radical mechanisms. Also, the possibilities for combined therapy including antioxidants in the acute and convalescent stages of stroke are considered. Future investigations are expected to elucidate the role of free radical processes in the chronic phase after stroke and to evaluate the prophylactic and therapeutic potential of anti-radical agents.
Collapse
Affiliation(s)
- Margarita L Alexandrova
- Department of Biophysics, Medical University, 1 Kliment Ohridsky str., 5800 Pleven, Bulgaria.
| | | |
Collapse
|
48
|
Brüx A, Girbes ARJ, Polderman KH. [Controlled mild-to-moderate hypothermia in the intensive care unit]. Anaesthesist 2005; 54:225-44. [PMID: 15742173 DOI: 10.1007/s00101-005-0808-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Controlled hypothermia is used as a therapeutic intervention to provide neuroprotection and (more recently) cardioprotection. The growing insight into the underlying pathophysiology of apoptosis and destructive processes at the cellular level, and the mechanisms underlying the protective effects of hypothermia, have led to improved application and to a widening of the range of potential indications. In many centres hypothermia has now become part of the standard therapy for post-anoxic coma in certain patients, but for other indications its use still remains controversial. The negative findings of some studies may be partly explained by inadequate protocols for the application of hypothermia and insufficient attention to the prevention of potential side effects. This review deals with some of the concepts underlying hypothermia-associated neuroprotection and cardioprotection, and discusses some potential clinical indications as well as reasons why some clinical trials may have produced conflicting results. Practical aspects such as methods to induce hypothermia, as well as the side effects of cooling are also discussed.
Collapse
Affiliation(s)
- A Brüx
- Abteilung Intensivmedizin, Freie Universität Medisch Centrum Amsterdam, Niederlande
| | | | | |
Collapse
|
49
|
Casas CE, Herrera LP, Prusmack C, Ruenes G, Marcillo A, Guest JD. Effects of epidural hypothermic saline infusion on locomotor outcome and tissue preservation after moderate thoracic spinal cord contusion in rats. J Neurosurg Spine 2005; 2:308-18. [PMID: 15796356 DOI: 10.3171/spi.2005.2.3.0308] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Object. Regionally delivered hypothermia has advantages over systemic hypothermia for clinical application following spinal cord injury (SCI). The effects of local hypothermia on tissue sparing, neuronal preservation, and locomotor outcome were studied in a moderate thoracic spinal cord contusion model.
Methods. Rats were randomized to four treatment groups and data were collected and analyzed in a blinded fashion. Chilled saline was perfused into the epidural space 30 minutes postcontusion to achieve the following epidural temperatures: 24 ± 2.3°C (16 rats), 30 ± 2.4°C (13 rats), and 35 ± 0.9°C (13 rats). Hypothermia was continued for 3 hours when a 45-minute period of rewarming was instituted. In a fourth group a moderate contusion only was induced in 14 animals. Rectal (core) and T9–10 (epidural) temperatures were measured continuously. Locomotor testing, using the Basso-Beattie-Bresnahan (Ba-Be-Br) scale, was performed for 6 weeks, and rats were videotaped for subsequent analysis. The lesion/preserved tissue ratio was calculated throughout the entire lesion cavity and the total lesion, spinal cord, and spared tissue volumes were determined. The rostral and caudal extent of gray matter loss was also measured. At 6 weeks locomotor recovery was similar in all groups (mean Ba-Be-Br Scale scores 14.88 ± 3.71, 14.83 ± 2.81, 14.50 ± 2.24, and 14.07 ± 2.39 [p = 0.77] for all four groups, respectively). No significant differences in spared tissue volumes were found when control and treatment groups were compared, but gray matter preservation was reduced in the infusion-treated groups.
Conclusions. Regional cooling applied 30 minutes after a moderate contusive SCI was not beneficial in terms of tissue sparing, neuronal preservation, or locomotor outcome. This method of cooling may reduce blood flow in the injured spinal cord and exacerbate secondary injury.
Collapse
Affiliation(s)
- Carlos E Casas
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Florida 33136, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Regulating a patient's body temperature has long been within the scope of practice of the critical care nurse. Different measures and modalities have been used to achieve normothermia in the past. Recent research has demonstrated how crucial body temperature can be, not only because of its potential for neuroprotection but also because of its effects on all body systems. The general consensus of current literature is that maintaining mild hypothermia at 32 degrees to 34 degrees C (89.6 degrees-93.2 degrees F) for 12 to 24 hours after cardiac arrest may provide optimal neuroprotection with minimal complications for patients.
Collapse
Affiliation(s)
- Marie Lasater
- Neurosurgery Intensive Care Unit, Barnes Jewish Hospital, One Barnes-Jewish Plaza, St. Louis, MO 63110, USA.
| |
Collapse
|