1
|
Barros M, Liang M, Iannucci N, Dickinson R. Xenon and Argon as Neuroprotective Treatments for Perinatal Hypoxic-Ischemic Brain Injury: A Preclinical Systematic Review and Meta-Analysis. Anesth Analg 2024:00000539-990000000-01012. [PMID: 39453983 DOI: 10.1213/ane.0000000000007223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Xenon and argon are currently being evaluated as potential neuroprotective treatments for acquired brain injuries. Xenon has been evaluated clinically as a treatment for brain ischemia with equivocal results in small trials, but argon has not yet undergone clinical evaluation. Several preclinical studies have investigated xenon or argon as treatments in animal models of perinatal hypoxic-ischemic encephalopathy (HIE). A systematic review of MEDLINE and Embase databases was performed. After screening of titles, abstracts, and full text, data were extracted from included studies. A pairwise meta-analysis of neuroprotective efficacy was performed using a random effects model. Heterogeneity was investigated using subgroup analysis, funnel plot asymmetry, and Egger's regression. The protocol was prospectively registered on PROSPERO (CRD42022301986). A total of 21 studies met the inclusion criteria. The data extracted included measurements from 1591 animals, involving models of HIE in mice, rats, and pigs. The meta-analysis found that both xenon and argon had significant (P < .0001) neuroprotective efficacies. The summary estimate for xenon was 39.7% (95% confidence interval [CI], 28.3%-51.1%) and for argon it was 70.3% (95% CI, 59.0%-81.7%). The summary effect for argon was significantly (P < .001) greater than that of xenon. Our results provide evidence supporting further investigation of xenon and argon as neuroprotective treatments for HIE.
Collapse
Affiliation(s)
- Mariana Barros
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Min Liang
- Anaesthesiology Research Institute, Department of Anaesthesiology, First Affiliated Hospital of Fujian Medical University, Binhai Campus, Fuzhou, China
| | - Noemi Iannucci
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Robert Dickinson
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Centre for Blast Injury Studies, Imperial College London, London, United Kingdom
| |
Collapse
|
2
|
Krenzlin H, Wesp DMA, Korinek AAE, Ubbens H, Volland J, Masomi-Bornwasser J, Weber KJ, Mole D, Sommer C, Ringel F, Alessandri B, Keric N. Effects of Argon in the Acute Phase of Subarachnoid Hemorrhage in an Endovascular Perforation Model in Rats. Neurocrit Care 2024:10.1007/s12028-024-02090-3. [PMID: 39174846 DOI: 10.1007/s12028-024-02090-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a devastating disease with high morbidity and mortality. Neuroprotective effects of the noble gas argon have been shown in animal models of ischemia. The aim of this study was to investigate the effects of argon in the immediate early phase of SAH in a rat model. METHODS A total of 19 male Wistar rats were randomly assigned to three treatment groups. SAH was induced using a endovascular filament perforation model. Cerebral blood flow, mean arterial blood pressure (MAP), and body temperature were measured continuously. Group A received 2 h of ventilation by 50% argon/50% O2 (n = 7) immediately following SAH. Group B underwent a sham operation and was also ventilated by 50% argon/50% O2 (n = 6). Group C underwent SAH and 50% O2/50% N2 ventilation (n = 6). Preoperative and postoperative neurological and behavioral testing were performed. Histology and immunohistochemistry were used to evaluate the extent of brain injury and vasospasm. RESULTS The cerebral blood flow dropped in both treatment groups after SAH induction (SAH, 63.0 ± 11.6% of baseline; SAH + argon, 80.2 ± 8.2% of baseline). During SAH, MAP increased (135.2 ± 10.5%) compared with baseline values (85.8 ± 26.0 mm Hg) and normalized thereafter. MAP in both groups showed no significant differences (p = 0.3123). Immunohistochemical staining for neuronal nuclear antigen demonstrated a decrease of hippocampal immunoreactivity after SAH in the cornu ammonis region (CA) 1-3 compared with baseline hippocampal immunoreactivity (p = 0.0127). Animals in the argon-ventilated group showed less neuronal loss compared with untreated SAH animals (p < 0.0001). Ionized calcium-binding adaptor molecule 1 staining showed a decreased accumulation after SAH + argon (CA1, 2.57 ± 2.35%; CA2, 1.89 ± 1.89%; CA3, 2.19 ± 1.99%; DG, 2.6 ± 2.24%) compared with untreated SAH animals (CA1, 5.48 ± 2.39%; CA2, 4.85 ± 4.06%; CA3, 4.22 ± 3.01%; dentate gyrus (DG), 3.82 ± 3.23%; p = 0.0007). The neuroscore assessment revealed no treatment benefit after SAH compared with baseline (p = 0.385). CONCLUSION In the present study, neuroprotective effects of argon occurred early after SAH. Because neurological deterioration was similar in the preadministration and absence of argon, it remains uncertain if neuroprotective effects translate in improved outcome over time.
Collapse
Affiliation(s)
- Harald Krenzlin
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Dominik M A Wesp
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Anika A E Korinek
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Henning Ubbens
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Jakob Volland
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Julia Masomi-Bornwasser
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Katharina J Weber
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dominik Mole
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Clemens Sommer
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Beat Alessandri
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Naureen Keric
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| |
Collapse
|
3
|
Bao L, Liu Y, Jia Q, Chu S, Jiang H, He S. Argon neuroprotection in ischemic stroke and its underlying mechanism. Brain Res Bull 2024; 212:110964. [PMID: 38670471 DOI: 10.1016/j.brainresbull.2024.110964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Ischemic stroke (IS), primarily caused by cerebrovascular obstruction, results in severe neurological deficits and has emerged as a leading cause of death and disability worldwide. Recently, there has been increasing exploration of the neuroprotective properties of the inert gas argon. Argon has exhibited impressive neuroprotection in many in vivo and ex vivo experiments without signs of adverse effects, coupled with the advantages of being inexpensive and easily available. However, the efficient administration strategy and underlying mechanisms of neuroprotection by argon in IS are still unclear. This review summarizes current research on the neuroprotective effects of argon in IS with the goal to provide effective guidance for argon application and to elucidate the potential mechanisms of argon neuroprotection. Early and appropriate argon administration at as high a concentration as possible offers favorable neuroprotection in IS. Argon inhalation has been shown to provide some long-term protection benefits. Argon provides the anti-oxidative stress, anti-inflammatory and anti-apoptotic cytoprotective effects mainly around Toll-like receptor 2/4 (TLR2/4), mediated by extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), nuclear factor kappa-B (NF-ĸB) and B-cell leukemia/lymphoma 2 (Bcl-2). Therefore, argon holds significant promise as a novel clinical neuroprotective gas agent for ischemic stroke after further researches to identify the optimal application strategy and elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Li Bao
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Yongxin Liu
- Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Qi Jia
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Sihao Chu
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Han Jiang
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Shuang He
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
4
|
Merigo G, Florio G, Madotto F, Magliocca A, Silvestri I, Fumagalli F, Cerrato M, Motta F, De Giorgio D, Panigada M, Zanella A, Grasselli G, Ristagno G. Treatment with inhaled Argon: a systematic review of pre-clinical and clinical studies with meta-analysis on neuroprotective effect. EBioMedicine 2024; 103:105143. [PMID: 38691938 PMCID: PMC11070688 DOI: 10.1016/j.ebiom.2024.105143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Argon (Ar) has been proposed as a potential therapeutic agent in multiple clinical conditions, specifically in organ protection. However, conflicting data on pre-clinical models, together with a great variability in Ar administration protocols and outcome assessments, have been reported. The aim of this study was to review evidence on treatment with Ar, with an extensive investigation on its neuroprotective effect, and to summarise all tested administration protocols. METHODS Using the PubMed database, all existing pre-clinical and clinical studies on the treatment with Ar were systematically reviewed (registration: https://doi.org/10.17605/OSF.IO/7983D). Study titles and abstracts were screened, extracting data from relevant studies post full-text review. Exclusion criteria included absence of full text and non-English language. Furthermore, meta-analysis was also performed to assess Ar potential as neuroprotectant agent in different clinical conditions: cardiac arrest, traumatic brain injury, ischemic stroke, perinatal hypoxic-ischemic encephalopathy, subarachnoid haemorrhage. Standardised mean differences for neurological, cognitive and locomotor, histological, and physiological measures were evaluated, through appropriate tests, clinical, and laboratory variables. In vivo studies were evaluated for risk of bias using the Systematic Review Center for Laboratory Animal Experimentation tool, while in vitro studies underwent assessment with a tool developed by the Office of Health Assessment and Translation. FINDINGS The systematic review detected 60 experimental studies (16 in vitro, 7 ex vivo, 31 in vivo, 6 with both in vitro and in vivo) investigating the role of Ar. Only one clinical study was found. Data from six in vitro and nineteen in vivo studies were included in the meta-analyses. In pre-clinical models, Ar administration resulted in improved neurological, cognitive and locomotor, and histological outcomes without any change in physiological parameters (i.e., absence of adverse events). INTERPRETATION This systematic review and meta-analysis based on experimental studies supports the neuroprotective effect of Ar, thus providing a rationale for potential translation of Ar treatment in humans. Despite adherence to established guidelines and methodologies, limitations in data availability prevented further analyses to investigate potential sources of heterogeneity due to study design. FUNDING This study was funded in part by Italian Ministry of Health-Current researchIRCCS and by Ministero della Salute Italiano, Ricerca Finalizzata, project no. RF 2019-12371416.
Collapse
Affiliation(s)
- Giulia Merigo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gaetano Florio
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Fabiana Madotto
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Aurora Magliocca
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Ivan Silvestri
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Francesca Fumagalli
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marianna Cerrato
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca Motta
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Daria De Giorgio
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mauro Panigada
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Zanella
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giacomo Grasselli
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Ristagno
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|
5
|
Wu D, Zhang D, Yin H, Zhang B, Xing J. Meta-analysis of the effects of inert gases on cerebral ischemia-reperfusion injury. Sci Rep 2023; 13:16896. [PMID: 37803128 PMCID: PMC10558482 DOI: 10.1038/s41598-023-43859-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 09/29/2023] [Indexed: 10/08/2023] Open
Abstract
Recently, noble gas has become a hot spot within the medical field like respiratory organ cerebral anemia, acute urinary organ injury and transplantation. However, the shield performance in cerebral ischemia-reperfusion injury (CIRI) has not reached an accord. This study aims to evaluate existing evidence through meta-analysis to determine the effects of inert gases on the level of blood glucose, partial pressure of oxygen, and lactate levels in CIRI. We searched relevant articles within the following both Chinese and English databases: PubMed, Web of science, Embase, CNKI, Cochrane Library and Scopus. The search was conducted from the time of database establishment to the end of May 2023, and two researchers independently entered the data into Revman 5.3 and Stata 15.1. There were total 14 articles were enclosed within the search. The results showed that the amount of partial pressure of blood oxygen in the noble gas cluster was beyond that in the medicine gas cluster (P < 0.05), and the inert gas group had lower lactate acid and blood glucose levels than the medical gas group. The partial pressure of oxygen (SMD = 1.51, 95% CI 0.10 ~ 0.91 P = 0.04), the blood glucose level (SMD = - 0.59, 95% CI - 0.92 ~ - 0.27 P = 0.0004) and the lactic acid level (SMD = - 0.42, 95% CI - 0.80 ~ - 0.03 P = 0.03) (P < 0.05). These results are evaluated as medium-quality evidence. Inert gas can effectively regulate blood glucose level, partial pressure of oxygen and lactate level, and this regulatory function mainly plays a protective role in the small animal ischemia-reperfusion injury model. This finding provides an assessment and evidence of the effectiveness of inert gases for clinical practice, and provides the possibility for the application of noble gases in the treatment of CIRI. However, more operations are still needed before designing clinical trials, such as the analysis of the inhalation time, inhalation dose and efficacy of different inert gases, and the effective comparison of the effects in large-scale animal experiments.
Collapse
Affiliation(s)
- Di Wu
- Department of Emergency Medicine, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Daoyu Zhang
- The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Hang Yin
- Baicheng Medical College, Baicheng, 137000, Jilin, China
| | - Bo Zhang
- The Second Foreign Department, Corps Hospital of the Chinese People's Armed Police Force of Jilin Province, Changchun, 130052, Jilin, China
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, No.71 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
6
|
Yin H, Chen Z, Zhao H, Huang H, Liu W. Noble gas and neuroprotection: From bench to bedside. Front Pharmacol 2022; 13:1028688. [PMID: 36532733 PMCID: PMC9750501 DOI: 10.3389/fphar.2022.1028688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/03/2022] [Indexed: 07/26/2023] Open
Abstract
In recent years, inert gases such as helium, argon, and xenon have gained considerable attention for their medical value. Noble gases present an intriguing scientific paradox: although extremely chemically inert, they display a remarkable spectrum of clinically useful biological properties. Despite a relative paucity of knowledge about their mechanisms of action, some noble gases have been used successfully in clinical practice. The neuroprotection elicited by these noble gases has been investigated in experimental animal models of various types of brain injuries, such as traumatic brain injury, stroke, subarachnoid hemorrhage, cerebral ischemic/reperfusion injury, and neurodegenerative diseases. Collectively, these central nervous system injuries are a leading cause of morbidity and mortality every year worldwide. Treatment options are presently limited to thrombolytic drugs and clot removal for ischemic stroke, or therapeutic cooling for other brain injuries before the application of noble gas. Currently, there is increasing interest in noble gases as novel treatments for various brain injuries. In recent years, neuroprotection elicited by particular noble gases, xenon, for example, has been reported under different conditions. In this article, we have reviewed the latest in vitro and in vivo experimental and clinical studies of the actions of xenon, argon, and helium, and discuss their potential use as neuroprotective agents.
Collapse
Affiliation(s)
- Haiying Yin
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zijun Chen
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hailin Zhao
- Division of Anesthetics, Department of Surgery and Cancer, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Han Huang
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wenwen Liu
- Department of Anesthesia Nursing, West China Second University Hospital, Sichuan University/West China School of Nursing, Ministry of Education, Sichuan University and Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, China
| |
Collapse
|
7
|
Scheid S, Lejarre A, Wollborn J, Buerkle H, Goebel U, Ulbrich F. Argon preconditioning protects neuronal cells with a Toll-like receptor-mediated effect. Neural Regen Res 2022; 18:1371-1377. [PMID: 36453425 PMCID: PMC9838174 DOI: 10.4103/1673-5374.355978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The noble gas argon has the potential to protect neuronal cells from cell death. So far, this effect has been studied in treatment after acute damage. Preconditioning using argon has not yet been investigated. In this study, human neuroblastoma SH-SY5Y cells were treated with different concentrations of argon (25%, 50%, and 74%; 21% O2, 5% CO2, balance nitrogen) at different time intervals before inflicting damage with rotenone (20 µM, 4 hours). Apoptosis was determined by flow cytometry after annexin V and propidium iodide staining. Surface expressions of Toll-like receptors 2 and 4 were also examined. Cells were also processed for analysis by western blot and qPCR to determine the expression of apoptotic and inflammatory proteins, such as extracellular-signal regulated kinase (ERK1/2), nuclear transcription factor-κB (NF-κB), protein kinase B (Akt), caspase-3, Bax, Bcl-2, interleukin-8, and heat shock proteins. Immunohistochemical staining was performed for TLR2 and 4 and interleukin-8. Cells were also pretreated with OxPAPC, an antagonist of TLR2 and 4 to elucidate the molecular mechanism. Results showed that argon preconditioning before rotenone application caused a dose-dependent but not a time-dependent reduction in the number of apoptotic cells. Preconditioning with 74% argon for 2 hours was used for further experiments showing the most promising results. Argon decreased the surface expression of TLR2 and 4, whereas OxPAPC treatment partially abolished the protective effect of argon. Argon increased phosphorylation of ERK1/2 but decreased NF-κB and Akt. Preconditioning inhibited mitochondrial apoptosis and the heat shock response. Argon also suppressed the expression of the pro-inflammatory cytokine interleukin-8. Immunohistochemistry confirmed the alteration of TLRs and interleukin-8. OxPAPC reversed the argon effect on ERK1/2, Bax, Bcl-2, caspase-3, and interleukin-8 expression, but not on NF-κB and the heat shock proteins. Taken together, argon preconditioning protects against apoptosis of neuronal cells and mediates its action via Toll-like receptors. Argon may represent a promising therapeutic alternative in various clinical settings, such as the treatment of stroke.
Collapse
Affiliation(s)
- Stefanie Scheid
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Adrien Lejarre
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Jakob Wollborn
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany,Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hartmut Buerkle
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Ulrich Goebel
- Department of Anesthesiology and Critical Care Medicine, St. Franziskus-Hospital, Muenster, Germany
| | - Felix Ulbrich
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany,Correspondence to: Felix Ulbrich, .
| |
Collapse
|
8
|
Liang M, Ahmad F, Dickinson R. Neuroprotection by the noble gases argon and xenon as treatments for acquired brain injury: a preclinical systematic review and meta-analysis. Br J Anaesth 2022; 129:200-218. [PMID: 35688658 PMCID: PMC9428918 DOI: 10.1016/j.bja.2022.04.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND The noble gases argon and xenon are potential novel neuroprotective treatments for acquired brain injuries. Xenon has already undergone early-stage clinical trials in the treatment of ischaemic brain injuries, with mixed results. Argon has yet to progress to clinical trials as a treatment for brain injury. Here, we aim to synthesise the results of preclinical studies evaluating argon and xenon as neuroprotective therapies for brain injuries. METHODS After a systematic review of the MEDLINE and Embase databases, we carried out a pairwise and stratified meta-analysis. Heterogeneity was examined by subgroup analysis, funnel plot asymmetry, and Egger's regression. RESULTS A total of 32 studies were identified, 14 for argon and 18 for xenon, involving measurements from 1384 animals, including murine, rat, and porcine models. Brain injury models included ischaemic brain injury after cardiac arrest (CA), neurological injury after cardiopulmonary bypass (CPB), traumatic brain injury (TBI), and ischaemic stroke. Both argon and xenon had significant (P<0.001), positive neuroprotective effect sizes. The overall effect size for argon (CA, TBI, stroke) was 18.1% (95% confidence interval [CI], 8.1-28.1%), and for xenon (CA, TBI, stroke) was 34.1% (95% CI, 24.7-43.6%). Including the CPB model, only present for xenon, the xenon effect size (CPB, CA, TBI, stroke) was 27.4% (95% CI, 11.5-43.3%). Xenon, both with and without the CPB model, was significantly (P<0.001) more protective than argon. CONCLUSIONS These findings provide evidence to support the use of xenon and argon as neuroprotective treatments for acquired brain injuries. Current evidence suggests that xenon is more efficacious than argon overall.
Collapse
Affiliation(s)
- Min Liang
- Anaesthetics, Pain Medicine, and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Fatin Ahmad
- Anaesthetics, Pain Medicine, and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Robert Dickinson
- Anaesthetics, Pain Medicine, and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, UK,Royal British Legion Centre for Blast Injury Studies, Imperial College London, London, UK,Corresponding author
| |
Collapse
|
9
|
He J, Xue K, Liu J, Gu JH, Peng B, Xu L, Wang G, Jiang Z, Li X, Zhang Y. Timely and Appropriate Administration of Inhaled Argon Provides Better Outcomes for tMCAO Mice: A Controlled, Randomized, and Double-Blind Animal Study. Neurocrit Care 2022; 37:91-101. [PMID: 35137354 DOI: 10.1007/s12028-022-01448-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inhaled argon (iAr) has shown promising therapeutic efficacy for acute ischemic stroke and has exhibited impressive advantages over other inert gases as a neuroprotective agent. However, the optimal dose, duration, and time point of iAr for acute ischemic stroke are unknown. Here, we explored variable iAr schedules and evaluated the neuroprotective effects of acute iAr administration on lesion volume, brain edema, and neurological function in a mouse model of cerebral ischemic/reperfusion injury. METHODS Adult ICR (Institute of Cancer Research) mice were randomly subjected to sham, moderate (1.5 h), or severe (3 h) transient middle cerebral artery occlusion (tMCAO). One hour after tMCAO, the mice were randomized to variable iAr protocols or air. General and focal deficit scores were assessed during double-blind treatment. Infarct volume, overall recovery, and brain edema were analyzed 24 h after cerebral ischemic/reperfusion injury. RESULTS Compared with those in the tMCAO-only group, lesion volume (p < 0.0001) and neurologic outcome (general, p < 0.0001; focal, p < 0.0001) were significantly improved in the group administered iAr 1 h after stroke onset (during ischemia). Short-term argon treatment (1 or 3 h) significantly improved the infarct volume (1 vs. 24 h, p < 0.0001; 3 vs. 24 h, p < 0.0001) compared with argon inhalation for 24 h. The concentration of iAr was confirmed to be a key factor in improving focal neurological outcomes relative to that in the tMCAO group, with higher concentrations of iAr showing better effects. Additionally, even though ischemia research has shown an increase in cerebral damage proportional to the ischemia time, argon administration showed significant neuroprotective effects on infarct volume (p < 0.0001), neurological deficits (general, p < 0.0001; focal, p < 0.0001), weight recovery (p < 0.0001), and edema (p < 0.0001) in general, particularly in moderate stroke. CONCLUSIONS Timely iAr administration during ischemia showed optimal neurological outcomes and minimal infarct volumes. Moreover, an appropriate duration of argon administration was important for better neuroprotective efficacy. These findings may provide vital guidance for using argon as a neuroprotective agent and moving to clinical trials in acute ischemic stroke.
Collapse
Affiliation(s)
- Juan He
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Ke Xue
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jiayi Liu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jin-Hua Gu
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China
| | - Bin Peng
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Lihua Xu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Guohua Wang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Zhenglin Jiang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Xia Li
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yunfeng Zhang
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China.
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
10
|
Antonova VV, Silachev DN, Ryzhkov IA, Lapin KN, Kalabushev SN, Ostrova IV, Varnakova LA, Grebenchikov OA. Three-Hour Argon Inhalation Has No Neuroprotective Effect after Open Traumatic Brain Injury in Rats. Brain Sci 2022; 12:brainsci12070920. [PMID: 35884727 PMCID: PMC9313057 DOI: 10.3390/brainsci12070920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/16/2022] Open
Abstract
In vivo studies of the therapeutic effects of argon in traumatic brain injury (TBI) are limited, and their results are contradictory. The aim of this study was to evaluate the effect of a three-hour inhalation of argon (70%Ar/30%O2) after an open TBI on the severity of the neurological deficit and the degree of brain damage in rats. The experiments were performed on male Wistar rats (n = 35). The TBI was simulated by the dosed open brain contusion injury. The animals were divided into three groups: sham-operated (SO, n = 7); TBI + 70%N2/30%O2 (TBI, n = 14); TBI + 70%Ar/30%O2 (TBI + iAr, n = 14). The Neurological status was assessed over a 14-day period (using the limb-placing and cylinder tests). Magnetic resonance imaging (MRI) scans and a histological examination of the brain with an assessment of the volume of the lesions were performed 14 days after the injury. At each of the time points (days 1, 7, and 14), the limb-placing test score was lower in the TBI and TBI + iAr groups than in the SO group, while there were no significant differences between the TBI and TBI + iAr groups. Additionally, no differences were found between these groups in the cylinder test scores (day 13). The volume of brain damage (tissue loss) according to both the MRI and histological findings did not differ between the TBI and TBI + iAr groups. A three-hour inhalation of argon (70%Ar/30%O2) after a TBI had no neuroprotective effect.
Collapse
Affiliation(s)
- Viktoriya V. Antonova
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
- Correspondence: ; Tel.: +7-938-500-3034
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - Ivan A. Ryzhkov
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
| | - Konstantin N. Lapin
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
| | - Sergey N. Kalabushev
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
- Institute of Functional Genomics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Irina V. Ostrova
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
| | - Lydia A. Varnakova
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
| | - Oleg A. Grebenchikov
- V.A. Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (I.A.R.); (K.N.L.); (S.N.K.); (I.V.O.); (L.A.V.); (O.A.G.)
| |
Collapse
|
11
|
Jin X, Li P, Michalski D, Li S, Zhang Y, Jolkkonen J, Cui L, Didwischus N, Xuan W, Boltze J. Perioperative stroke: A perspective on challenges and opportunities for experimental treatment and diagnostic strategies. CNS Neurosci Ther 2022; 28:497-509. [PMID: 35224865 PMCID: PMC8928912 DOI: 10.1111/cns.13816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023] Open
Abstract
Perioperative stroke is an ischemic or hemorrhagic cerebral event during or up to 30 days after surgery. It is a feared condition due to a relatively high incidence, difficulties in timely detection, and unfavorable outcome compared to spontaneously occurring stroke. Recent preclinical data suggest that specific pathophysiological mechanisms such as aggravated neuroinflammation contribute to the detrimental impact of perioperative stroke. Conventional treatment options are limited in the perioperative setting due to difficult diagnosis and medications affecting coagulation in may cases. On the contrary, the chance to anticipate cerebrovascular events at the time of surgery may pave the way for prevention strategies. This review provides an overview on perioperative stroke incidence, related problems, and underlying pathophysiological mechanisms. Based on this analysis, we assess experimental stroke treatments including neuroprotective approaches, cell therapies, and conditioning medicine strategies regarding their potential use in perioperative stroke. Interestingly, the specific aspects of perioperative stroke might enable a more effective application of experimental treatment strategies such as classical neuroprotection whereas others including cell therapies may be of limited use. We also discuss experimental diagnostic options for perioperative stroke augmenting classical clinical and imaging stroke diagnosis. While some experimental stroke treatments may have specific advantages in perioperative stroke, the paucity of established guidelines or multicenter clinical research initiatives currently limits their thorough investigation.
Collapse
Affiliation(s)
- Xia Jin
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | | | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yueman Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Jukka Jolkkonen
- Department of Neurology and A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lili Cui
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Nadine Didwischus
- School of Life Sciences, University of Warwick, Coventry, UK.,Department of Radiology, University of Pittsburgh, Pittsburgh, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
12
|
Zhang J, Liu W, Bi M, Xu J, Yang H, Zhang Y. Noble Gases Therapy in Cardiocerebrovascular Diseases: The Novel Stars? Front Cardiovasc Med 2022; 9:802783. [PMID: 35369316 PMCID: PMC8966230 DOI: 10.3389/fcvm.2022.802783] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiocerebrovascular diseases (CCVDs) are the leading cause of death worldwide; therefore, to deeply explore the pathogenesis of CCVDs and to find the cheap and efficient strategies to prevent and treat CCVDs, these are of great clinical and social significance. The discovery of nitric oxide (NO), as one of the endothelium-derived relaxing factors and its successful utilization in clinical practice for CCVDs, provides new ideas for us to develop drugs for CCVDs: “gas medicine” or “medical gases.” The endogenous gas molecules such as carbon monoxide (CO), hydrogen sulfide (H2S), sulfur dioxide (SO2), methane (CH4), and hydrogen (H2) have essential biological effects on modulating cardiocerebrovascular homeostasis and CCVDs. Moreover, it has been shown that noble gas atoms such as helium (He), neon (Ne), argon (Ar), krypton (Kr), and xenon (Xe) display strong cytoprotective effects and therefore, act as the exogenous pharmacologic preventive and therapeutic agents for CCVDs. Mechanistically, besides the competitive inhibition of N-methyl-D-aspartate (NMDA) receptor in nervous system by xenon, the key and common mechanisms of noble gases are involved in modulation of cell death and inflammatory or immune signals. Moreover, gases interaction and reduction in oxidative stress are emerging as the novel biological mechanisms of noble gases. Therefore, to investigate the precise actions of noble gases on redox signals, gases interaction, different cell death forms, and the emerging field of gasoimmunology, which focus on the effects of gas atoms/molecules on innate immune signaling or immune cells under both the homeostatic and perturbed conditions, these will help us to uncover the mystery of noble gases in modulating CCVDs.
Collapse
Affiliation(s)
- Jiongshan Zhang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Institute of Integrated Traditional Chinese and Western Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wei Liu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- Research Centre for Integrative Medicine (Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research), Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingmin Bi
- Department of Otorhinolaryngology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinwen Xu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- Research Centre for Integrative Medicine (Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research), Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongzhi Yang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Institute of Integrated Traditional Chinese and Western Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaxing Zhang
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- Research Centre for Integrative Medicine (Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research), Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
13
|
Schneider FI, Krieg SM, Lindauer U, Stoffel M, Ryang YM. Neuroprotective Effects of the Inert Gas Argon on Experimental Traumatic Brain Injury In Vivo with the Controlled Cortical Impact Model in Mice. BIOLOGY 2022; 11:biology11020158. [PMID: 35205025 PMCID: PMC8869506 DOI: 10.3390/biology11020158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Traumatic brain injuries remain one of the leading causes of death in the western world and developing countries. There is an urgent need for causal therapies for such injuries. The noble gas argon has already shown promising results in in-vitro models. The influence of argon on the extent of damage after a craniocerebral trauma will be investigated in this study, in vivo, in mice. After the trauma, the animals were examined for neurological impairments and their brains were removed to detect brain edema and microscopically detectable alterations. Abstract Argon has shown neuroprotective effects after traumatic brain injury (TBI) and cerebral ischemia in vitro and in focal cerebral ischemia in vivo. The purpose of this study is to show whether argon beneficially impacts brain contusion volume (BCV) as the primary outcome parameter, as well as secondary outcome parameters, such as brain edema, intracranial pressure (ICP), neurological outcome, and cerebral blood flow (CBF) in an in-vivo model. Subjects were randomly assigned to either argon treatment or room air. After applying controlled cortical impact (CCI) onto the dura with 8 m/s (displacement 1 mm, impact duration 150 ms), treatment was administered by a recovery chamber with 25%, 50%, or 75% argon and the rest being oxygen for 4 h after trauma. Two control groups received room air for 15 min and 24 h, respectively. Neurological testing and ICP measurements were performed 24 h after trauma, and brains were removed to measure secondary brain damage. The primary outcome parameter, BCV, and the secondary outcome parameter, brain edema, were not significantly reduced by argon treatment at any concentration. There was a highly significant decrease in ICP at 50% argon (p = 0.001), and significant neurological improvement (beamwalk missteps) at 25% and 50% argon (p = 0.01; p = 0.049 respectively) compared to control.
Collapse
Affiliation(s)
- Fritz I. Schneider
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany; (F.I.S.); (U.L.); (M.S.); (Y.-M.R.)
| | - Sandro M. Krieg
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany; (F.I.S.); (U.L.); (M.S.); (Y.-M.R.)
- Correspondence: ; Tel.: +49-89-4140-2151
| | - Ute Lindauer
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany; (F.I.S.); (U.L.); (M.S.); (Y.-M.R.)
- Department of Neurosurgery, Klinikum der RWTH Aachen, RWTH Aachen University, 52062 Aachen, Germany
| | - Michael Stoffel
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany; (F.I.S.); (U.L.); (M.S.); (Y.-M.R.)
- Department of Neurosurgery, Helios Kliniken, 47805 Krefeld, Germany
| | - Yu-Mi Ryang
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany; (F.I.S.); (U.L.); (M.S.); (Y.-M.R.)
| |
Collapse
|
14
|
Post-stroke treatment with argon preserved neurons and attenuated microglia/macrophage activation long-termly in a rat model of transient middle cerebral artery occlusion (tMCAO). Sci Rep 2022; 12:691. [PMID: 35027642 PMCID: PMC8758662 DOI: 10.1038/s41598-021-04666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 12/24/2021] [Indexed: 11/18/2022] Open
Abstract
In a previous study from our group, argon has shown to significantly attenuate brain injury, reduce brain inflammation and enhance M2 microglia/macrophage polarization until 7 days after ischemic stroke. However, the long-term effects of argon have not been reported thus far. In the present study, we analyzed the underlying neuroprotective effects and potential mechanisms of argon, up to 30 days after ischemic stroke. Argon administration with a 3 h delay after stroke onset and 1 h after reperfusion demonstrated long-term neuroprotective effect by preserving the neurons at the ischemic boundary zone 30 days after stroke. Furthermore, the excessive microglia/macrophage activation in rat brain was reduced by argon treatment 30 days after ischemic insult. However, long-lasting neurological improvement was not detectable. More sensorimotor functional measures, age- and disease-related models, as well as further histological and molecular biological analyses will be needed to extend the understanding of argon’s neuroprotective effects and mechanism of action after ischemic stroke.
Collapse
|
15
|
Kremer B, Coburn M, Weinandy A, Nolte K, Clusmann H, Veldeman M, Höllig A. Argon treatment after experimental subarachnoid hemorrhage: evaluation of microglial activation and neuronal survival as a subanalysis of a randomized controlled animal trial. Med Gas Res 2021; 10:103-109. [PMID: 33004706 PMCID: PMC8086619 DOI: 10.4103/2045-9912.296039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hereinafter, we evaluate argon's neuroprotective and immunomodulatory properties after experimental subarachnoid hemorrhage (SAH) examining various localizations (hippocampal and cortical regions) with respect to neuronal damage and microglial activation 6, 24 and 72 hours after SAH. One hour after SAH (endovascular perforation rat model) or sham surgery, a mixture of gas containing 50% argon (argon group) or 50% nitrogen (control group) was applied for 1 hour. At 6 hours after SAH, argon reduced neuronal damage in the hippocampal regions in the argon group compared to the control group (P < 0.034). Hippocampal microglial activation did not differ between the treatment groups over time. The basal cortical regions did not show a different lesion pattern, but microglial activation was significantly reduced in the argon group 72 hours after SAH (P = 0.034 vs. control group). Whereas callosal microglial activation was significantly reduced at 24 hours in the argon-treated group (P = 0.018). Argon treatment ameliorated only early hippocampal neuronal damage after SAH. Inhibition of microglial activation was seen in some areas later on. Thus, argon may influence the microglial inflammatory response and neuronal survival after SAH; however, due to low sample sizes the interpretation of our results is limited. The study protocol was approved by the Government Agency for Animal Use and Protection (Protocol number: TVA 10416G1; initially approved by the "Landesamt für Natur, Umwelt und Verbraucherschutz NRW," Recklinghausen, Germany, on April 28, 2009).
Collapse
Affiliation(s)
- Benedikt Kremer
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Mark Coburn
- Department of Anaesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Agnieszka Weinandy
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Kay Nolte
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Hans Clusmann
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Michael Veldeman
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
16
|
Creed J, Cantillana-Riquelme V, Yan BH, Ma S, Chu D, Wang H, Turner DA, Laskowitz DT, Hoffmann U. Argon Inhalation for 24 h After Closed-Head Injury Does not Improve Recovery, Neuroinflammation, or Neurologic Outcome in Mice. Neurocrit Care 2021; 34:833-843. [PMID: 32959200 DOI: 10.1007/s12028-020-01104-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND/OBJECTIVE In recent years, the noble gas argon (Ar) has been extensively studied for its organ protection properties. While mounting in vitro and in vivo evidence indicates that argon provides neuroprotection in ischemic brain injury, its neuroprotective potential in traumatic brain injury (TBI) has not been evaluated in vivo. We tested the hypothesis that prolonged inhalation of 70% or 79% argon for 24 h after closed-head injury (CHI) improves neurologic outcome and overall recovery at 36 days post-injury. We also compared effects of the 30% or 21% residual oxygen on argon's potential neuroprotective capacity. METHODS Adult male C57/black mice (n = 240) were subjected to closed-head traumatic brain injury, followed by inhalation of 70% argon or nitrogen (30% oxygen), or 79% argon or nitrogen (21% oxygen) for 24 h. Neurologic outcome (rotarod, neuroscore, and Morris water maze) was evaluated for up to 36 days post-injury. Histologic parameters of neurologic degeneration (Fluoro-Jade staining) and inflammation (F4/80 microglia immunostaining) were assessed in subgroups at 24 h and on post-injury day 7. RESULTS Our CHI protocol consistently resulted in significant brain injury. After argon inhalation for 24 h at either concentration, mice did not show significant improvement with regard to neuroscores, rotarod performance, Morris water maze performance, or overall recovery (body weight), compared to nitrogen controls, up to 36 days. At 7 days post-injury, histologic markers of neurodegeneration and inflammation, particularly in the hippocampus, consistently demonstrated significant injury. Notably, recovery was reduced in mice treated with the higher oxygen concentration (30%) after CHI compared to 21%. CONCLUSIONS Prolonged argon treatment did not improve neurologic outcome, overall recovery (weight), nor markers of neurodegeneration or neuroinflammation after significant CHI compared to nitrogen. While neuroprotective in predominately ischemic injury, argon did not provide protection after TBI in this model, highlighting the crucial importance of assessing argon's strengths and weaknesses in preclinical models to fully understand its organ protective potential in different pathologies and gas mixtures.
Collapse
Affiliation(s)
- Jennifer Creed
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | | | - Bai Hui Yan
- Department of Anesthesiology, Center for Perioperative Organ Protection (CPOP), Duke University Medical Center, Box 3094, Durham, NC, 27710, USA
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an JiaoTong University, Xi'an, ShaanXi Province, China
| | - Shuang Ma
- Department of Anesthesiology, Center for Perioperative Organ Protection (CPOP), Duke University Medical Center, Box 3094, Durham, NC, 27710, USA
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liao Ning, China
| | - Dongmei Chu
- Department of Anesthesiology, Center for Perioperative Organ Protection (CPOP), Duke University Medical Center, Box 3094, Durham, NC, 27710, USA
- Department of Pediatrics, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Haichen Wang
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Dennis A Turner
- Department of Anesthesiology, Center for Perioperative Organ Protection (CPOP), Duke University Medical Center, Box 3094, Durham, NC, 27710, USA
- Departments of Neurosurgery, Neurobiology, and Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | - Daniel T Laskowitz
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Department of Anesthesiology, Center for Perioperative Organ Protection (CPOP), Duke University Medical Center, Box 3094, Durham, NC, 27710, USA
| | - Ulrike Hoffmann
- Department of Anesthesiology, Center for Perioperative Organ Protection (CPOP), Duke University Medical Center, Box 3094, Durham, NC, 27710, USA.
| |
Collapse
|
17
|
Inhaled gases as novel neuroprotective therapies in the postcardiac arrest period. Curr Opin Crit Care 2021; 27:255-260. [PMID: 33769417 DOI: 10.1097/mcc.0000000000000820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize recent advances about inhaled gases as novel neuroprotective agents in the postcardiac arrest period. RECENT FINDINGS Inhaled gases, as nitric oxide (NO) and molecular hydrogen (H2), and noble gases as xenon (Xe) and argon (Ar) have shown neuroprotective properties after resuscitation. In experimental settings, the protective effect of these gases has been demonstrated in both in-vitro studies and animal models of cardiac arrest. They attenuate neuronal degeneration and improve neurological function after resuscitation acting on different pathophysiological pathways. Safety of both Xe and H2 after cardiac arrest has been reported in phase 1 clinical trials. A randomized phase 2 clinical trial showed the neuroprotective effects of Xe, combined with targeted temperature management. Xe inhalation for 24 h after resuscitation preserves white matter integrity as measured by fractional anisotropy of diffusion tensor MRI. SUMMARY Inhaled gases, as Xe, Ar, NO, and H2 have consistently shown neuroprotective effects in experimental studies. Ventilation with these gases appears to be well tolerated in pigs and in preliminary human trials. Results from phase 2 and 3 clinical trials are needed to assess their efficacy in the treatment of postcardiac arrest brain injury.
Collapse
|
18
|
Argon Attenuates Multiorgan Failure in Relation with HMGB1 Inhibition. Int J Mol Sci 2021; 22:ijms22063257. [PMID: 33806919 PMCID: PMC8111890 DOI: 10.3390/ijms22063257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 11/17/2022] Open
Abstract
Argon inhalation attenuates multiorgan failure (MOF) after experimental ischemic injury. We hypothesized that this protection could involve decreased High Mobility Group Box 1 (HMGB1) systemic release. We investigated this issue in an animal model of MOF induced by aortic cross-clamping. Anesthetized rabbits were submitted to supra-coeliac aortic cross-clamping for 30 min, followed by 300 min of reperfusion. They were randomly divided into three groups (n = 7/group). The Control group inhaled nitrogen (70%) and oxygen (30%). The Argon group was exposed to a mixture of argon (70%) and oxygen (30%). The last group inhaled nitrogen/oxygen (70/30%) with an administration of the HMGB1 inhibitor glycyrrhizin (4 mg/kg i.v.) 5 min before aortic unclamping. At the end of follow-up, cardiac output was significantly higher in Argon and Glycyrrhizin vs. Control (60 ± 4 and 49 ± 4 vs. 33 ± 8 mL/kg/min, respectively). Metabolic acidosis was attenuated in Argon and Glycyrrhizin vs. Control, along with reduced amount of norepinephrine to reverse arterial hypotension. This was associated with reduced interleukin-6 and HMGB1 plasma concentration in Argon and Glycyrrhizin vs. Control. End-organ damages were also attenuated in the liver and kidney in Argon and Glycyrrhizin vs. Control, respectively. Argon inhalation reduced HMGB1 blood level after experimental aortic cross-clamping and provided similar benefits to direct HMGB1 inhibition.
Collapse
|
19
|
Goebel U, Scheid S, Spassov S, Schallner N, Wollborn J, Buerkle H, Ulbrich F. Argon reduces microglial activation and inflammatory cytokine expression in retinal ischemia/reperfusion injury. Neural Regen Res 2021; 16:192-198. [PMID: 32788476 PMCID: PMC7818862 DOI: 10.4103/1673-5374.290098] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We previously found that argon exerts its neuroprotective effect in part by inhibition of the toll-like receptors (TLR) 2 and 4. The downstream transcription factors signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-κB) are also affected by argon and may play a role in neuroprotection. It also has been demonstrated that argon treatment could mitigate brain damage, reduce excessive microglial activation, and subsequently attenuate brain inflammation. Despite intensive research, the further exact mechanism remains unclear. In this study, human neuroblastoma cells were damaged in vitro with rotenone over a period of 4 hours (to mimic cerebral ischemia and reperfusion damage), followed by a 2-hour post-conditioning with argon (75%). In a separate in vivo experiment, retinal ischemia/reperfusion injury was induced in rats by increasing intraocular pressure for 1 hour. Upon reperfusion, argon was administered by inhalation for 2 hours. Argon reduced the binding of the transcription factors signal transducer and activator of transcription 3, nuclear factor kappa B, activator protein 1, and nuclear factor erythroid 2-related factor 2, which are involved in regulation of neuronal damage. Flow cytometry analysis showed that argon downregulated the Fas ligand. Some transcription factors were regulated by toll-like receptors; therefore, their effects could be eliminated, at least in part, by the TLR2 and TLR4 inhibitor oxidized phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC). Argon treatment reduced microglial activation after retinal ischemia/reperfusion injury. Subsequent quantitative polymerase chain reaction analysis revealed a reduction in the pro-inflammatory cytokines interleukin (IL-1α), IL-1β, IL-6, tumor necrosis factor α, and inducible nitric oxide synthase. Our results suggest that argon reduced the extent of inflammation in retinal neurons after ischemia/reperfusion injury by suppression of transcription factors crucial for microglial activation. Argon has no known side effects or narcotic properties; therefore, therapeutic use of this noble gas appears ideal for treatment of patients with neuronal damage in retinal ischemia/reperfusion injury. The animal experiments were approved by the Commission for Animal Care of the University of Freiburg (approval No. 35-9185.81/G14-122) on October 19, 2012.
Collapse
Affiliation(s)
- Ulrich Goebel
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefanie Scheid
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sashko Spassov
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jakob Wollborn
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Buerkle
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Ulbrich
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
20
|
Argon: a noble, but not inert, treatment for brain trauma? Br J Anaesth 2020; 126:41-43. [PMID: 33097180 DOI: 10.1016/j.bja.2020.09.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/01/2023] Open
|
21
|
Inhaled Argon Impedes Hepatic Regeneration after Ischemia/Reperfusion Injury in Rats. Int J Mol Sci 2020; 21:ijms21155457. [PMID: 32751707 PMCID: PMC7432339 DOI: 10.3390/ijms21155457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 11/16/2022] Open
Abstract
Organoprotective effects of noble gases are subject of current research. One important field of interest is the effect of noble gases on hepatic regenerative capacity. For the noble gas argon, promising studies demonstrated remarkable experimental effects in neuronal and renal cells. The aim of this study was to investigate the effects of argon on the regenerative capacity of the liver after ischemia/reperfusion injury (IRI). Male, Sprague-Dawley rats underwent hepatic IRI by clamping of the hepatic artery. Expression of hepatoproliferative genes (HGF, IL-1β, IL-6, TNF), cell cycle markers (BrdU, TUNEL, Ki-67), and liver enzymes (ALT, AST, Bilirubin, LDH) were assessed 3, 36, and 96 h after IRI. Expression of IL-1β and IL-6 was significantly higher after argon inhalation after 36 h (IL-1β 5.0 vs. 8.7 fold, p = 0.001; IL-6 9.6 vs. 19.1 fold, p = 0.05). Ki-67 was higher in the control group compared to the argon group after 36 h (214.0 vs. 38.7 positive cells/1000 hepatocytes, p = 0.045). Serum levels of AST and ALT did not differ significantly between groups. Our data indicate that argon inhalation has detrimental effects on liver regeneration after IRI as measured by elevated levels of the proinflammatory cytokines IL-1β and IL-6 after 36 h. In line with these results, Ki-67 is decreased in the argon group, indicating a negative effect on liver regeneration in argon inhalation.
Collapse
|
22
|
Nespoli F, Redaelli S, Ruggeri L, Fumagalli F, Olivari D, Ristagno G. A complete review of preclinical and clinical uses of the noble gas argon: Evidence of safety and protection. Ann Card Anaesth 2020; 22:122-135. [PMID: 30971592 PMCID: PMC6489383 DOI: 10.4103/aca.aca_111_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The noble gas argon (Ar) is a "biologically" active element and has been extensively studied preclinically for its organ protection properties. This work reviews all preclinical studies employing Ar and describes the clinical uses reported in literature, analyzing 55 pertinent articles found by means of a search on PubMed and Embase. Ventilation with Ar has been tested in different models of acute disease at concentrations ranging from 20% to 80% and for durations between a few minutes up to days. Overall, lesser cell death, smaller infarct size, and better functional recovery after ischemia have been repeatedly observed. Modulation of the molecular pathways involved in cell survival, with resulting anti-apoptotic and pro-survival effects, appeared as the determinant mechanism by which Ar fulfills its protective role. These beneficial effects have been reported regardless of onset and duration of Ar exposure, especially after cardiac arrest. In addition, ventilation with Ar was safe both in animals and humans. Thus, preclinical and clinical data support future clinical studies on the role of inhalatory Ar as an organ protector.
Collapse
Affiliation(s)
- Francesca Nespoli
- Department of Cardiovascular Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Simone Redaelli
- Department of Cardiovascular Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Ruggeri
- Department of Cardiovascular Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca Fumagalli
- Department of Cardiovascular Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Davide Olivari
- Department of Cardiovascular Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giuseppe Ristagno
- Department of Cardiovascular Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
23
|
Argon Inhalation for 24 Hours After Onset of Permanent Focal Cerebral Ischemia in Rats Provides Neuroprotection and Improves Neurologic Outcome. Crit Care Med 2020; 47:e693-e699. [PMID: 31094741 DOI: 10.1097/ccm.0000000000003809] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES We tested the hypothesis that prolonged inhalation of 70% argon for 24 hours after in vivo permanent or temporary stroke provides neuroprotection and improves neurologic outcome and overall recovery after 7 days. DESIGN Controlled, randomized, double-blinded laboratory study. SETTING Animal research laboratories. SUBJECTS Adult Wistar male rats (n = 110). INTERVENTIONS Rats were subjected to permanent or temporary focal cerebral ischemia via middle cerebral artery occlusion, followed by inhalation of 70% argon or nitrogen in 30% oxygen for 24 hours. On postoperative day 7, a 48-point neuroscore and histologic lesion size were assessed. MEASUREMENTS AND MAIN RESULTS After argon inhalation for 24 hours immediately following "severe permanent ischemia" induction, neurologic outcome (neuroscore, p = 0.034), overall recovery (body weight, p = 0.02), and infarct volume (total infarct volume, p = 0.0001; cortical infarct volume, p = 0.0003; subcortical infarct volume, p = 0.0001) were significantly improved. When 24-hour argon treatment was delayed for 2 hours after permanent stroke induction or until after postischemic reperfusion treatment, neurologic outcomes remained significantly improved (neuroscore, p = 0.043 and p = 0.014, respectively), as was overall recovery (body weight, p = 0.015), compared with nitrogen treatment. However, infarct volume and 7-day mortality were not significantly reduced when argon treatment was delayed. CONCLUSIONS Neurologic outcome (neuroscore), overall recovery (body weight), and infarct volumes were significantly improved after 24-hour inhalation of 70% argon administered immediately after severe permanent stroke induction. Neurologic outcome and overall recovery were also significantly improved even when argon treatment was delayed for 2 hours or until after reperfusion.
Collapse
|
24
|
Anna R, Rolf R, Mark C. Update of the organoprotective properties of xenon and argon: from bench to beside. Intensive Care Med Exp 2020; 8:11. [PMID: 32096000 PMCID: PMC7040108 DOI: 10.1186/s40635-020-0294-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
The growth of the elderly population has led to an increase in patients with myocardial infarction and stroke (Wajngarten and Silva, Eur Cardiol 14: 111–115, 2019). Patients receiving treatment for ST-segment-elevation myocardial infarction (STEMI) highly profit from early reperfusion therapy under 3 h from the onset of symptoms. However, mortality from STEMI remains high due to the increase in age and comorbidities (Menees et al., N Engl J Med 369: 901–909, 2013). These factors also account for patients with acute ischaemic stroke. Reperfusion therapy has been established as the gold standard within the first 4 to 5 h after onset of symptoms (Powers et al., Stroke 49: e46-e110, 2018). Nonetheless, not all patients are eligible for reperfusion therapy. The same is true for traumatic brain injury patients. Due to the complexity of acute myocardial and central nervous injury (CNS), finding organ protective substances to improve the function of remote myocardium and the ischaemic penumbra of the brain is urgent. This narrative review focuses on the noble gases argon and xenon and their possible cardiac, renal and neuroprotectant properties in the elderly high-risk (surgical) population. The article will provide an overview of the latest experimental and clinical studies. It is beyond the scope of this review to give a detailed summary of the mechanistic understanding of organ protection by xenon and argon.
Collapse
Affiliation(s)
- Roehl Anna
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52072, Aachen, Germany.
| | - Rossaint Rolf
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52072, Aachen, Germany
| | - Coburn Mark
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52072, Aachen, Germany
| |
Collapse
|
25
|
Pretreatment With Argon Protects Human Cardiac Myocyte-Like Progenitor Cells from Oxygen Glucose Deprivation-Induced Cell Death by Activation of AKT and Differential Regulation of Mapkinases. Shock 2019; 49:556-563. [PMID: 29658909 DOI: 10.1097/shk.0000000000000998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The noble gas argon induces cardioprotection in a rabbit model of myocardial ischemia and reperfusion. However, no studies in human primary cells or subjects have been performed so far. We used human cardiac myocyte-like progenitor cells (HCMs) to investigate the protective effect on the cellular level. METHODS HCMs were pretreated with 30% or 50% argon before oxygen-glucose deprivation (OGD) and reperfusion. We evaluated apoptotic states by flow cytometry and the activation of mitogen-activated protein kinase (MAPKs) members extracellular signal-regulated kinase (ERK), c-jun N-terminal kinase (JNK), p38 MAPkinase, and protein kinase B (Akt) by Westernblot analysis and by activity assays of downstream transcription factors. Specific inhibitors were used to proof a significant participation of these pathways in the protection by argon. Beneficial effects were further assessed by TdT-mediated dUTP-biotin nick end labeling (TUNEL) assay, lactate dehydrogenase (LDH), mitochondrial deoxyribonucleic acid (mtDNA), and cytokine release. RESULTS Pretreatment with 30% or 50% argon for 90 min before OGD resulted in a significant protection of HCMs against apoptosis. This effect was reversed by the application of MAPK and Akt inhibitors during argon exposure. Argon 30% reduced the release of LDH by 33% and mtDNA by 45%. The release of interleukin 1β was reduced by 44% after OGD and more than 90% during reperfusion. CONCLUSIONS Pretreatment with argon protects HCMs from apoptosis under ischemic conditions via activation of Akt, Erk, and biphasic regulation of JNK. Argon gas is cheap and easily administrable, and might be a novel therapy to reduce myocardial ischemia-reperfusion injury.
Collapse
|
26
|
Liu J, Nolte K, Brook G, Liebenstund L, Weinandy A, Höllig A, Veldeman M, Willuweit A, Langen KJ, Rossaint R, Coburn M. Post-stroke treatment with argon attenuated brain injury, reduced brain inflammation and enhanced M2 microglia/macrophage polarization: a randomized controlled animal study. Crit Care 2019; 23:198. [PMID: 31159847 PMCID: PMC6547472 DOI: 10.1186/s13054-019-2493-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In recent years, argon has been shown to exert neuroprotective effects in an array of models. However, the mechanisms by which argon exerts its neuroprotective characteristics remain unclear. Accumulating evidence imply that argon may exert neuroprotective effects via modulating the activation and polarization of microglia/macrophages after ischemic stroke. In the present study, we analyzed the underlying neuroprotective effects of delayed argon application until 7 days after reperfusion and explored the potential mechanisms. METHODS Twenty-one male Wistar rats underwent transient middle cerebral artery occlusion or sham surgery randomly for 2 h using the endoluminal thread model. Three hours after transient middle cerebral artery occlusion induction and 1 h after reperfusion, animals received either 50% vol Argon/50% vol O2 or 50% vol N2/50% vol O2 for 1 h. The primary outcome was the 6-point neuroscore from 24 h to d7 after reperfusion. Histological analyses including infarct volume, survival of neurons (NeuN) at the ischemic boundary zone, white matter integrity (Luxol Fast Blue), microglia/macrophage activation (Iba1), and polarization (Iba1/Arginase1 double staining) on d7 were conducted as well. Sample size calculation was performed using nQuery Advisor + nTerim 4.0. Independent t test, one-way ANOVA and repeated measures ANOVA were performed, respectively, for statistical analysis (SPSS 23.0). RESULTS The 6-point neuroscore from 24 h to d7 after reperfusion showed that tMCAO Ar group displayed significantly improved neurological performance compared to tMCAO N2 group (p = 0.026). The relative numbers of NeuN-positive cells in the ROIs of tMCAO Ar group significantly increased compared to tMCAO N2 group (p = 0.010 for cortex and p = 0.011 for subcortex). Argon significantly suppressed the microglia/macrophage activation as revealed by Iba1 staining (p = 0.0076) and promoted the M2 microglia/macrophage polarization as revealed by Iba1/Arginase 1 double staining (p = 0.000095). CONCLUSIONS Argon administration with a 3 h delay after stroke onset and 1 h after reperfusion significantly alleviated neurological deficit within the first week and preserved the neurons at the ischemic boundary zone 7 days after stroke. Moreover, argon reduced the excessive microglia/macrophage activation and promoted the switch of microglia/macrophage polarization towards the anti-inflammatory M2 phenotype. Studies making efforts to further elucidate the protective mechanisms and to benefit the translational application are of great value.
Collapse
Affiliation(s)
- Jingjin Liu
- Department of Anesthesiology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Kay Nolte
- Department of Neuropathology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Gary Brook
- Department of Neuropathology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Lisa Liebenstund
- Department of Anesthesiology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Agnieszka Weinandy
- Department of Neuropathology, Medical Faculty RWTH Aachen University, Aachen, Germany
- Department of Neurosurgery, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Anke Höllig
- Department of Anesthesiology, Medical Faculty RWTH Aachen University, Aachen, Germany
- Department of Neurosurgery, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Michael Veldeman
- Department of Anesthesiology, Medical Faculty RWTH Aachen University, Aachen, Germany
- Department of Neurosurgery, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-4), Forschungszentrum Jülich, Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-4), Forschungszentrum Jülich, Jülich, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, Medical Faculty RWTH Aachen University, Aachen, Germany
| |
Collapse
|
27
|
Kiss A, Shu H, Hamza O, Santer D, Tretter EV, Yao S, Markstaller K, Hallström S, Podesser BK, Klein KU. Argon preconditioning enhances postischaemic cardiac functional recovery following cardioplegic arrest and global cold ischaemia. Eur J Cardiothorac Surg 2019; 54:539-546. [PMID: 29547976 DOI: 10.1093/ejcts/ezy104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 02/09/2018] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Previous studies demonstrated that preconditioning with argon gas provided a marked reduction in inflammation and apoptosis and increased myocardial contractility in the setting of acute myocardial ischaemia-reperfusion (IR). There is substantial evidence that myocardial IR injury following cardioplegic arrest is associated with the enhancement of apoptosis and inflammation, which is considered to play a role in cardiac functional impairment. Therefore, the present study was designed to clarify whether preconditioning with argon gas enhances recovery of cardiac function following cardioplegic arrest. METHODS Sprague-Dawley rats were anaesthetized and ventilated and allocated to (i) the control group (control IR, n = 10) and (ii) the in vivo group (argon IR), which received 3 cycles of argon (50% argon, 21% oxygen and 29% nitrogen, n = 10) administered for 5 min interspersed with 5 min of a gas mixture (79% nitrogen and 21% oxygen). The hearts were excised and then evaluated in an erythrocyte-perfused isolated working heart system. Cold ischaemia (4°C) for 60 min was induced by histidine-tryptophan-ketoglutarate cardioplegia, followed by 40 min of reperfusion. Cardiac functional parameters were assessed. In left ventricular tissue samples, the expressions of extracellular-regulated kinase (ERK1/2), AKT serine/threonine kinase (Akt), jun N-terminal kinase (JNK), endothelial nitric oxide synthase (eNOS) and HMGB1: high-mobility group box 1 (HMGB1) protein were assessed by western blot, and high-energy phosphates were evaluated by high-performance liquid chromatography. RESULTS At the end of reperfusion, the rats preconditioned with argon showed significantly enhanced recovery of cardiac output (101 ± 6% vs 87 ± 11%; P < 0.01), stroke volume (94 ± 4% vs 80 ± 11%; P = 0.001), external heart work (100 ± 6% vs 81 ± 13%; P < 0.001) and coronary flow (90 ± 13% vs 125 ± 21%; P < 0.01) compared with the control IR group. These results were accompanied by a significant increase in the levels of myocardial phosphocreatine (23.71 ± 2.07 µmol/g protein vs the control IR group, 13.50 ± 4.75; P = 0.001) and maintained adenosine triphosphate levels (13.62 ±1.89 µmol/g protein vs control IR group adenosine triphosphate: 10.08 ± 1.94 µmol/g; P = 0.017). Additionally, preconditioning with argon markedly reduced the activation of JNK (0.11 ± 0.01 vs 0.25 ± 0.03; P = 0.005) and the expression of HMGB1 protein (0.52 ± 0.04 vs 1.5 ± 0.10; P < 0.001) following reperfusion. CONCLUSIONS Preconditioning with argon enhanced cardiac functional recovery in rat hearts arrested with histidine-tryptophan-ketoglutarate cardioplegia, thereby representing a potential novel cardioprotective approach in cardiac surgery.
Collapse
Affiliation(s)
- Attila Kiss
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Huaqing Shu
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria.,Department of Anesthesiology and Intensive Care Unit, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ouafa Hamza
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - David Santer
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University of Vienna, Vienna, Austria.,Department of Cardiovascular Surgery, Hospital Hietzing, Vienna, Austria
| | - Eva Verena Tretter
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Shanglong Yao
- Department of Anesthesiology and Intensive Care Unit, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Klaus Markstaller
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Seth Hallström
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University of Vienna, Vienna, Austria.,Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University Graz, Graz, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Klaus Ulrich Klein
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Suleiman S, Klassen S, Katz I, Balakirski G, Krabbe J, von Stillfried S, Kintsler S, Braunschweig T, Babendreyer A, Spillner J, Kalverkamp S, Schröder T, Moeller M, Coburn M, Uhlig S, Martin C, Rieg AD. Argon reduces the pulmonary vascular tone in rats and humans by GABA-receptor activation. Sci Rep 2019; 9:1902. [PMID: 30760775 PMCID: PMC6374423 DOI: 10.1038/s41598-018-38267-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Argon exerts neuroprotection. Thus, it might improve patients' neurological outcome after cerebral disorders or cardiopulmonary resuscitation. However, limited data are available concerning its effect on pulmonary vessel and airways. We used rat isolated perfused lungs (IPL) and precision-cut lung slices (PCLS) of rats and humans to assess this topic. IPL: Airway and perfusion parameters, oedema formation and the pulmonary capillary pressure (Pcap) were measured and the precapillary and postcapillary resistance (Rpost) was calculated. In IPLs and PCLS, the pulmonary vessel tone was enhanced with ET-1 or remained unchanged. IPLs were ventilated and PCLS were gassed with argon-mixture or room-air. IPL: Argon reduced the ET-1-induced increase of Pcap, Rpost and oedema formation (p < 0.05). PCLS (rat): Argon relaxed naïve pulmonary arteries (PAs) (p < 0.05). PCLS (rat/human): Argon attenuated the ET-1-induced contraction in PAs (p < 0.05). Inhibition of GABAB-receptors abolished argon-induced relaxation (p < 0.05) in naïve or ET-1-pre-contracted PAs; whereas inhibition of GABAA-receptors only affected ET-1-pre-contracted PAs (p < 0.01). GABAA/B-receptor agonists attenuated ET-1-induced contraction in PAs and baclofen (GABAB-agonist) even in pulmonary veins (p < 0.001). PLCS (rat): Argon did not affect the airways. Finally, argon decreases the pulmonary vessel tone by activation of GABA-receptors. Hence, argon might be applicable in patients with pulmonary hypertension and right ventricular failure.
Collapse
Affiliation(s)
- Said Suleiman
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Sergej Klassen
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Ira Katz
- Medical Research & Development, Air Liquide Santé Internationale, Centre de Recherche Paris-Saclay, 78354, Jouy-en-Josas, France
| | - Galina Balakirski
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Julia Krabbe
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | | | - Svetlana Kintsler
- Institute of Pathology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Till Braunschweig
- Institute of Pathology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Jan Spillner
- Department of Cardiac and Thoracic Surgery, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Sebastian Kalverkamp
- Department of Cardiac and Thoracic Surgery, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Thomas Schröder
- Department of Surgery, Luisenhospital Aachen, 52064, Aachen, Germany
| | - Manfred Moeller
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Mark Coburn
- Department of Anaesthesiology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Annette D Rieg
- Department of Anaesthesiology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany.
| |
Collapse
|
29
|
Ning J, Zhao H, Chen B, Mi EZ, Yang Z, Qing W, Lam KWJ, Yi B, Chen Q, Gu J, Ichim T, Bogin V, Lu K, Ma D. Argon Mitigates Impaired Wound Healing Process and Enhances Wound Healing In Vitro and In Vivo. Theranostics 2019; 9:477-490. [PMID: 30809288 PMCID: PMC6376177 DOI: 10.7150/thno.29361] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/04/2018] [Indexed: 12/26/2022] Open
Abstract
Diabetic foot ulcers are associated with significant morbidity and mortality, and current treatments are far from optimal. Chronic wounds in diabetes are characterised by impaired angiogenesis, leukocyte function, fibroblast proliferation, and keratinocyte migration and proliferation. Methods: We tested the effect of exposure to argon gas on endothelial cell, fibroblast, macrophage and keratinocyte cell cultures in vitro and in vivo of a streptozotocin-induced diabetic mouse model. Results: Exposure to normobaric argon gas promotes multiple steps of the wound healing process. Argon accelerated angiogenesis, associated with upregulation of pro-angiogenic Angiopoietin-1 and vascular endothelial growth factor (VEGF) signalling in vitro and in vivo. Treatment with argon enhanced expression of transforming growth factor (TGF)-β, early recruitment of macrophages and keratinocyte proliferation. Argon had a pro-survival effect, inducing expression of cytoprotective mediators B-cell lymphoma 2 and heme oxygenase 1. Argon was able to accelerate wound closure in a diabetic mouse model. Conclusion: Together these findings indicate that argon gas may be a promising candidate for clinical use in treatment of diabetic ulcers.
Collapse
|
30
|
Tolaymat Y, Doré S, Griffin HW, Shih S, Edwards ME, Weiss MD. Inhaled Gases for Neuroprotection of Neonates: A Review. Front Pediatr 2019; 7:558. [PMID: 32047729 PMCID: PMC6996209 DOI: 10.3389/fped.2019.00558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/20/2019] [Indexed: 11/30/2022] Open
Abstract
Importance: Hypoxic-ischemic encephalopathy (HIE) is a significant cause of morbidity and mortality in neonates. The incidence of HIE is 1-8 per 1,000 live births in developed countries. Whole-body hypothermia reduces the risk of disability or death, but 7 infants needed to be treated to prevent death or major neurodevelopmental disability. Inhalational gases may be promising synergistic agents due to their rapid onset and easy titratability. Objective: To review current data on different inhaled gases with neuroprotective properties that may serve as adjunct therapies to hypothermia. Evidence review: Literature review was performed using the PubMed database, google scholar, and ClinicalTrials.Gov. Results focused on articles published from January 1, 2005, through December 31, 2017. Articles published earlier than 2005 were included when appropriate for historical perspective. Our review emphasized preclinical and clinical studies relevant to the use of inhaled agents for neuroprotection. Findings: Based on the relevance to our topic, 111 articles were selected pertaining to the incidence of HIE, pathophysiology of HIE, therapeutic hypothermia, and emerging therapies for hypoxic-ischemic encephalopathy in preclinical and clinical settings. Supplemental tables summarizes highly relevant 49 publications that were included in this review. The selected publications emphasize the emergence of promising inhaled gases that may improve neurologic survival and alleviate neurodevelopmental disability when combined with therapeutic hypothermia in the future. Conclusions: Many inhaled agents have neuroprotective properties and could serve as an adjunct therapy to whole-body hypothermia. Inhaled agents are ideal due to their easy administration, titrability, and rapid onset and offset.
Collapse
Affiliation(s)
- Youness Tolaymat
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Sylvain Doré
- Departments of Neurology, Psychiatry, Pharmaceuticals and Neuroscience, University of Florida, Gainesville, FL, United States
| | - Hudson W Griffin
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Susana Shih
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Mary E Edwards
- Health Science Center Libraries, University of Florida, Gainesville, FL, United States
| | - Michael D Weiss
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
31
|
|
32
|
Abstract
Stroke, also known as “cerebrovascular accident”, is an acute cerebrovascular disease that is caused by a sudden rupture of blood vessels in the brain or obstruction of the blood supply by blockage of blood vessels, thus including hemorrhagic and ischemic strokes. The incidence of ischemic stroke is higher than that of hemorrhagic stroke, and accounts for 80% of the total number of strokes. However, the mortality rate of hemorrhagic stroke is relatively high. Internal carotid artery and vertebral artery occlusion and stenosis can cause ischemic stroke, and especially males over 40 years of age are at a high risk of morbidity. According to the survey, stroke in urban and rural areas has become the first cause of death in China. It is also the leading cause of disability in Chinese adults. In a word, stroke is characterized by high morbidity, high mortality and high disability rates. Studies have shown that many noble gases have the neuroprotective effects. For example, xenon has been extensively studied in various animal models of neurological injury including stroke, hypoxic-ischemic encephalopathy. Compared to xenon, Argon, as a noble gas, is abundant, cheap and widely applicable, and has been also demonstrated to be neuroprotective in many research studies. In a variety of models, ranging from oxygen-glucose deprivation in cell culture to complex models of mid-cerebral artery occlusion, subarachnoid hemorrhage or retinal ischemia-reperfusion injury in animals. Argon administration after individual injury demonstrated favorable effects, particularly increased cell survival and even improved neuronal function. Therefore the neuroprotective effects of argon may be of possible clinical use for opening a potential therapeutic window in stroke. It is important to illuminate the mechanisms of argon in nerve function and to explore the best use of this gas in stroke treatment.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhu-Wei Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jin-Quan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Survival with favorable neurological function after cardiac arrest remains low. The purpose of this review is to identify recent advances that focus on neuroprotection during cardiopulmonary resuscitation (CPR). RECENT FINDINGS Multiple strategies have been shown to enhance neuroprotection during CPR. Brain perfusion during CPR is increased with therapies such as active compression decompression CPR and intrathoracic pressure regulation that improve cardiac preload and decrease intracranial pressure. Head Up CPR has been shown to decrease intracranial pressure thereby increasing cerebral perfusion pressure and cerebral blood flow. Sodium nitroprusside enhanced CPR increases cerebral perfusion, facilitates heat exchange, and improves neurologic survival in swine after cardiac arrest. Postconditioning has been administered during CPR in laboratory settings. Poloxamer 188, a membrane stabilizer, and ischemic postconditioning have been shown to improve cardiac and neural function after cardiac arrest in animal models. Postconditioning with inhaled gases protects the myocardium, with more evidence mounting for the potential for neural protection. SUMMARY Multiple promising neuroprotective therapies are being developed in animal models of cardiac arrest, and are in early stages of human trials. These therapies have the potential to be bundled together to improve rates of favorable neurological survival after cardiac arrest.
Collapse
|
34
|
Savary G, Lidouren F, Rambaud J, Kohlhauer M, Hauet T, Bruneval P, Costes B, Cariou A, Ghaleh B, Mongardon N, Tissier R. Argon attenuates multiorgan failure following experimental aortic cross-clamping. Br J Clin Pharmacol 2018; 84:1170-1179. [PMID: 29388238 DOI: 10.1111/bcp.13535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 12/27/2017] [Accepted: 01/16/2018] [Indexed: 12/16/2022] Open
Abstract
AIMS Argon has been shown to prevent ischaemic injuries in several scenarios of regional ischaemia. We determined whether it could provide a systemic effect in a model of multiorgan failure (MOF) induced by aortic cross-clamping. METHODS Anaesthetized rabbits were submitted to aortic cross-clamping (30 min) and subsequent reperfusion (300 min). They were either ventilated with oxygen-enriched air throughout the protocol [fraction of inspired oxygen (FiO2 ) = 30%; control group) or with a mixture of 30% oxygen and 70% argon (argon groups). In a first group treated with argon ('Argon-Total'), its administration was started 30 min before ischaemia and maintained throughout the protocol. In the two other groups, the administration was started either 30 min before ischaemia ('Argon-Pre') or at the onset of reperfusion ('Argon-Post'), for a total duration of 2 h. Cardiovascular, renal and inflammatory endpoints were assessed throughout protocol. RESULTS Compared with control, shock was significantly attenuated in Argon-Total and Argon-Pre but not Argon-Post groups (e.g. cardiac output = 62±5 vs. 29 ± 5 ml min-1 kg-1 in Argon-Total and control groups at the end of the follow-up). Shock and renal failure were reduced in all argon vs. control groups. Histopathological examination of the gut showed attenuation of ischaemic lesions in all argon vs. control groups. Blood transcription levels of interleukin (IL) 1β, IL-8, IL-10 and hypoxia-inducible factor 1α were not significantly different between groups. CONCLUSION Argon attenuated clinical and biological modifications of cardiovascular, renal and intestinal systems, but not the inflammatory response, after aortic cross-clamping. The window of administration was crucial to optimize organ protection.
Collapse
Affiliation(s)
- Guillaume Savary
- Inserm, U955, Equipe 3, Créteil, France.,Université Paris Est, UMR_S955, UPEC, DHU A-TVB Créteil, France.,Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | - Jérôme Rambaud
- Inserm, U955, Equipe 3, Créteil, France.,Université Paris Est, UMR_S955, UPEC, DHU A-TVB Créteil, France.,Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Matthias Kohlhauer
- Inserm, U955, Equipe 3, Créteil, France.,Université Paris Est, UMR_S955, UPEC, DHU A-TVB Créteil, France.,Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Thierry Hauet
- Faculté de Médecine et de Pharmacie, Inserm, U1082, Université de Poitiers, Poitiers, France.,Service de Biochimie, CHU de Poitiers, Poitiers, France
| | - Patrick Bruneval
- Service d'Anatomie Pathologique, Hôpital Européen Georges Pompidou, Assistance Publique des Hôpitaux de Paris, Paris, France
| | | | - Alain Cariou
- Service de Réanimation Médicale, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Bijan Ghaleh
- Inserm, U955, Equipe 3, Créteil, France.,Université Paris Est, UMR_S955, UPEC, DHU A-TVB Créteil, France.,Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Nicolas Mongardon
- Inserm, U955, Equipe 3, Créteil, France.,Université Paris Est, UMR_S955, UPEC, DHU A-TVB Créteil, France.,Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.,Service d'Anesthésie et des Réanimations Chirurgicales, DHU A-TVB, Hôpitaux Universitaires Henri Mondor, Assistance Publique des Hôpitaux de Paris, Créteil, France
| | - Renaud Tissier
- Inserm, U955, Equipe 3, Créteil, France.,Université Paris Est, UMR_S955, UPEC, DHU A-TVB Créteil, France.,Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| |
Collapse
|
35
|
Gardner A, Menon D. Moving to human trials for argon neuroprotection in neurological injury: a narrative review. Br J Anaesth 2018; 120:453-468. [DOI: 10.1016/j.bja.2017.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 12/12/2022] Open
|
36
|
|
37
|
Veldeman M, Coburn M, Rossaint R, Clusmann H, Nolte K, Kremer B, Höllig A. Xenon Reduces Neuronal Hippocampal Damage and Alters the Pattern of Microglial Activation after Experimental Subarachnoid Hemorrhage: A Randomized Controlled Animal Trial. Front Neurol 2017; 8:511. [PMID: 29021779 PMCID: PMC5623683 DOI: 10.3389/fneur.2017.00511] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/13/2017] [Indexed: 01/03/2023] Open
Abstract
Objective The neuroprotective properties of the noble gas xenon have already been demonstrated using a variety of injury models. Here, we examine for the first time xenon’s possible effect in attenuating early brain injury (EBI) and its influence on posthemorrhagic microglial neuroinflammation in an in vivo rat model of subarachnoid hemorrhage (SAH). Methods Sprague-Dawley rats (n = 22) were randomly assigned to receive either Sham surgery (n = 9; divided into two groups) or SAH induction via endovascular perforation (n = 13, divided into two groups). Of those randomized for SAH, 7 animals were postoperatively ventilated with 50 vol% oxygen/50 vol% xenon for 1 h and 6 received 50 vol% oxygen/50 vol% nitrogen (control). The animals were sacrificed 24 h after SAH. Of each animal, a cerebral coronal section (−3.60 mm from bregma) was selected for assessment of histological damage 24 h after SAH. A 5-point neurohistopathological severity score was applied to assess neuronal cell damage in H&E and NeuN stained sections in a total of four predefined anatomical regions of interest. Microglial activation was evaluated by a software-assisted cell count of Iba-1 stained slices in three cortical regions of interest. Results A diffuse cellular damage was apparent in all regions of the ipsilateral hippocampus 24 h after SAH. Xenon-treated animals presented with a milder damage after SAH. This effect was found to be particularly pronounced in the medial regions of the hippocampus, CA3 (p = 0.040), and dentate gyrus (DG p = 0.040). However, for the CA1 and CA2 regions, there were no statistical differences in neuronal damage according to our histological scoring. A cell count of activated microglia was lower in the cortex of xenon-treated animals. This difference was especially apparent in the left piriform cortex (p = 0.017). Conclusion In animals treated with 50 vol% xenon (for 1 h) after SAH, a less pronounced neuronal damage was observed for the ipsilateral hippocampal regions CA3 and DG, when compared to the control group. In xenon-treated animals, a lower microglial cell count was observed suggesting an immunomodulatory effect generated by xenon. As for now, these results cannot be generalized as only some hippocampal regions are affected. Future studies should assess the time and localization dependency of xenon’s beneficial properties after SAH.
Collapse
Affiliation(s)
- Michael Veldeman
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany.,Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Hans Clusmann
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Kay Nolte
- Department of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Benedikt Kremer
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
38
|
Grüßer L, Blaumeiser-Debarry R, Krings M, Kremer B, Höllig A, Rossaint R, Coburn M. Argon attenuates the emergence of secondary injury after traumatic brain injury within a 2-hour incubation period compared to desflurane: an in vitro study. Med Gas Res 2017; 7:93-100. [PMID: 28744361 PMCID: PMC5510299 DOI: 10.4103/2045-9912.208512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite years of research, treatment of traumatic brain injury (TBI) remains challenging. Considerable data exists that some volatile anesthetics might be neuroprotective. However, several studies have also revealed a rather neurotoxic profile of anesthetics. In this study, we investigated the effects of argon 50%, desflurane 6% and their combination in an in vitro TBI model with incubation times similar to narcotic time slots in a daily clinical routine. Organotypic hippocampal brain slices of 5- to 7-day-old mice were cultivated for 14 days before TBI was performed. Slices were eventually incubated for 2 hours in an atmosphere containing no anesthetic gas, argon 50% or desflurane 6% or both. Trauma intensity was evaluated via fluorescent imagery. Our results show that neither argon 50% nor desflurane 6% nor their combination could significantly reduce the trauma intensity in comparison to the standard atmosphere. However, in comparison to desflurane 6%, argon 50% displayed a rather neuroprotective profile within the first 2 hours after a focal mechanical trauma (P = 0.015). A 2-hour incubation in an atmosphere containing both gases, argon 50% and desflurane 6%, did not result in significant effects in comparison to the argon 50% group or the desflurane 6% group. Our findings demonstrate that within a 2-hour incubation time neither argon nor desflurane could affect propidium iodide-detectable cell death in an in vitro TBI model in comparison to the standard atmosphere, although cell death was less with argon 50% than with desflurane 6%. The results show that within this short time period processes concerning the development of secondary injury are already taking place and may be manipulated by argon.
Collapse
Affiliation(s)
- Linda Grüßer
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Matthias Krings
- Department of Anesthesiology and Intensive Care, Medizinisches Zentrum StaedteRegion Aachen, Wuerselen, Germany
| | - Benedikt Kremer
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
39
|
Litwin PD, Reis Dib AL, Chen J, Noga M, Finlay WH, Martin AR. Theoretical and experimental evaluation of the effects of an argon gas mixture on the pressure drop through adult tracheobronchial airway replicas. J Biomech 2017; 58:217-221. [DOI: 10.1016/j.jbiomech.2017.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/04/2017] [Accepted: 04/09/2017] [Indexed: 11/27/2022]
|
40
|
Influence of argon on temperature modulation and neurological outcome in hypothermia treated rats following cardiac arrest. Resuscitation 2017; 117:32-39. [PMID: 28579371 DOI: 10.1016/j.resuscitation.2017.05.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/24/2017] [Accepted: 05/29/2017] [Indexed: 11/21/2022]
Abstract
AIM OF THE STUDY Combining xenon and mild therapeutic hypothermia (MTH) after cardiac arrest (CA) confers a degree of protection that is greater than either of the two interventions alone. However, xenon is very costly which might preclude a widespread use. We investigated whether the inexpensive gas argon would enhance hypothermia induced neurologic recovery in a similar manner. METHODS Following nine minutes of CA and three minutes of cardiopulmonary resuscitation 21 male Sprague-Dawley rats were randomized to receive MTH (33°C for 6h), MTH plus argon (70% for 1h), or no treatment. A first day condition score assessed behaviour, motor activity and overall condition. A neurological deficit score (NDS) was calculated daily for seven days following the experiment before the animals were killed and the brains harvested for histopathological analysis. RESULTS All animals survived. Animals that received MTH alone showed best overall neurologic function. Strikingly, this effect was abolished in the argon-augmented MTH group, where animals showed worse neurologic outcome being significant in the first day condition score and on day one to three and five in the NDS in comparison to MTH treated rats. Results were reflected by the neurohistopathological analysis. CONCLUSION Our study demonstrates that argon augmented MTH does not improve functional recovery after CA in rats, but may even worsen neurologic function in this model.
Collapse
|
41
|
Modulation by the Noble Gas Helium of Tissue Plasminogen Activator: Effects in a Rat Model of Thromboembolic Stroke. Crit Care Med 2017; 44:e383-9. [PMID: 26646461 DOI: 10.1097/ccm.0000000000001424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTERVENTIONS Helium has been shown to provide neuroprotection in mechanical model of acute ischemic stroke by inducing hypothermia, a condition shown by itself to reduce the thrombolytic and proteolytic properties of tissue plasminogen activator. However, whether or not helium interacts with the thrombolytic drug tissue plasminogen activator, the only approved therapy of acute ischemic stroke still remains unknown. This point is not trivial since previous data have shown the critical importance of the time at which the neuroprotective noble gases xenon and argon should be administered, during or after ischemia, in order not to block tissue plasminogen activator-induced thrombolysis and to obtain neuroprotection and inhibition of tissue plasminogen activator-induced brain hemorrhages. MEASUREMENTS AND MAIN RESULTS We show that helium of 25-75 vol% inhibits in a concentration-dependent fashion the catalytic and thrombolytic activity of tissue plasminogen activator in vitro and ex vivo. In vivo, in rats subjected to thromboembolic brain ischemia, we found that intraischemic helium at 75 vol% inhibits tissue plasminogen activator-induced thrombolysis and subsequent reduction of ischemic brain damage and that postischemic helium at 75 vol% reduces ischemic brain damage and brain hemorrhages. CONCLUSIONS In a clinical perspective for the treatment of acute ischemic stroke, these data suggest that helium 1) should not be administered before or together with tissue plasminogen activator therapy due to the risk of inhibiting the benefit of tissue plasminogen activator-induced thrombolysis; and 2) could be an efficient neuroprotective agent if given after tissue plasminogen activator-induced reperfusion.
Collapse
|
42
|
Ulmer TF, Fragoulis A, Dohmeier H, Kroh A, Andert A, Stoppe C, Alizai H, Klink C, Coburn M, Neumann UP. Argon Delays Initiation of Liver Regeneration after Partial Hepatectomy in Rats. Eur Surg Res 2017; 58:204-215. [PMID: 28433997 DOI: 10.1159/000466690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The liver can heal up to restitutio ad integrum following damage resulting from various causes. Different studies have demonstrated the protective effect of argon on various cells and organs. To the best of our knowledge, the organ-protective effects of the noble gas argon on the liver have not yet been investigated, although argon appears to influence signal paths that are well-known mediators of liver regeneration. We hypothesized that argon inhalation prior to partial hepatectomy (70%) has a positive effect on the initiation of liver regeneration in rats. METHODS Partial hepatectomy (70%) with or without inhaled argon (50 vol%) was performed for 1 h. Liver tissue was harvested after 3, 36, and 96 h to analyze the mRNA and protein expression of hepatocyte growth factor (HGF), interleukin-6 (IL-6), tumor necrosis factor-α, and extracellular signal-regulated kinase 1/2. Histological tissue samples were prepared for immunohistochemistry (bromodeoxyuridine [BrdU], Ki-67, and TUNEL) and blood was analyzed regarding the effects of argon on liver function. Statistical analyses were performed using 1-way ANOVA followed by the post hoc Tukey-Kramer test. RESULTS After 3 h, the primary outcome parameter of hepatocyte proliferation was significantly reduced with argon 50 vol% inhalation in comparison to nitrogen inhalation (BrdU: 15.7 ± 9.7 vs. 7.7 ± 3.1 positive cells/1,000 hepatocytes, p = 0.013; Ki-67: 17.6 ± 13.3 vs. 4.7 ± 5.4 positive cells/1,000 hepatocytes, p = 0.006). This was most likely mediated by significant downregulation of HGF (after 3 h: 5.2 ± 3.2 vs. 2.3 ± 1.0 fold, p = 0.032; after 96 h: 2.1 ± 0.5 vs. 1.3 ± 0.3 fold, p = 0.029) and IL-6 (after 3 h: 43.7 ± 39.6 vs. 8.5 ± 9.2 fold, p = 0.032). Nevertheless, we could detect no significant effect on the weight of the residual liver, liver-body weight ratio, or liver blood test results after argon inhalation. CONCLUSION Impairment of liver regeneration was apparent after argon 50 vol% inhalation that was most probably mediated by downregulation of HGF and IL-6 in the initial phase. However, the present study was not adequately powered to prove that argon has detrimental effects on the liver. Further studies are needed to evaluate the effects of argon on livers with preexisting conditions as well as on ischemia-reperfusion models.
Collapse
Affiliation(s)
- Tom Florian Ulmer
- Department of General, Visceral, and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Martens A, Ordies S, Vanaudenaerde BM, Verleden SE, Vos R, Verleden GM, Verbeken EK, Van Raemdonck DE, Claes S, Schols D, Chalopin M, Katz I, Farjot G, Neyrinck AP. A porcine ex vivo lung perfusion model with maximal argon exposure to attenuate ischemia-reperfusion injury. Med Gas Res 2017; 7:28-36. [PMID: 28480029 PMCID: PMC5402344 DOI: 10.4103/2045-9912.202907] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Argon (Ar) is a noble gas with known organoprotective effects in rodents and in vitro models. In a previous study we failed to find a postconditioning effect of Ar during ex vivo lung perfusion (EVLP) on warm-ischemic injury in a porcine model. In this study, we further investigated a prolonged exposure to Ar to decrease cold ischemia-reperfusion injury after lung transplantation in a porcine model with EVLP assessment. Domestic pigs (n = 6/group) were pre-conditioned for 6 hours with 21% O2 and 79% N2 (CONTR) or 79% Ar (ARG). Subsequently, lungs were cold flushed and stored inflated on ice for 18 hours inflated with the same gas mixtures. Next, lungs were perfused for 4 hours on EVLP (acellular) while ventilated with 12% O2 and 88% N2 (CONTR group) or 88% Ar (ARG group). The perfusate was saturated with the same gas mixture but with the addition of CO2 to an end-tidal CO2 of 35-45 mmHg. The saturated perfusate was drained and lungs were perfused with whole blood for an additional 2 hours on EVLP. Evaluation at the end of EVLP did not show significant effects on physiologic parameters by prolonged exposure to Ar. Also wet-to-dry weight ratio did not improve in the ARG group. Although in other organ systems protective effects of Ar have been shown, we did not detect beneficial effects of a high concentration of Ar on cold pulmonary ischemia-reperfusion injury in a porcine lung model after prolonged exposure to Ar in this porcine model with EVLP assessment.
Collapse
Affiliation(s)
- An Martens
- Laboratory of Anesthesiology and Algology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium.,Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sofie Ordies
- Laboratory of Anesthesiology and Algology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium.,Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Pneumology, Department of Clinical and Experimental Medicine, Lung Transplant Unit, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium
| | - Stijn E Verleden
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Pneumology, Department of Clinical and Experimental Medicine, Lung Transplant Unit, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium
| | - Robin Vos
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Pneumology, Department of Clinical and Experimental Medicine, Lung Transplant Unit, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Pneumology, Department of Clinical and Experimental Medicine, Lung Transplant Unit, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium
| | - Eric K Verbeken
- University Hospitals Leuven, Department of Histopathology, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium
| | - Sandra Claes
- Laboratory of Virology and Chemotherapy (Rega Institute), Department of Microbiology and Immunology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy (Rega Institute), Department of Microbiology and Immunology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthieu Chalopin
- Air Liquide Santé International Medical R&D; Paris-Saclay Research Center, Jouy-en Josas, France
| | - Ira Katz
- Air Liquide Santé International Medical R&D; Paris-Saclay Research Center, Jouy-en Josas, France
| | - Geraldine Farjot
- Air Liquide Santé International Medical R&D; Paris-Saclay Research Center, Jouy-en Josas, France
| | - Arne P Neyrinck
- Laboratory of Anesthesiology and Algology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven and University Hospitals, Leuven, Belgium.,Leuven Lung Transplant Unit, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
44
|
Yıldız EP, Ekici B, Tatlı B. Neonatal hypoxic ischemic encephalopathy: an update on disease pathogenesis and treatment. Expert Rev Neurother 2016; 17:449-459. [PMID: 27830959 DOI: 10.1080/14737175.2017.1259567] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Hypoxic ischemic encephalopathy (HIE) is the most important reason for morbidity and mortality in term-born infants. Understanding pathophysiology of the brain damage is essential for the early detection of patients with high risk for HIE and development of strategies for their treatments. Areas covered: This review discusses pathophysiology of the neonatal HIE and its treatment options, including hypothermia, melatonin, allopurinol, topiramate, erythropoietin, N-acetylcyctein, magnesium sulphate and xenon. Expert commentary: Several clinical studies have been performed in order to decrease the risk of brain injury due to difficulties in the early diagnosis and treatment, and to develop strategies for better long-term outcomes. Although currently standard treatment methods include therapeutic hypothermia for neonates with moderate to severe HIE, new supportive options are needed to enhance neuroprotective effects of the hypothermia, which should aim to reduce production of the free radicals and to have anti-inflammatory and anti-apoptotic actions.
Collapse
Affiliation(s)
| | - Barış Ekici
- b Department of Pediatric Neurology , Liv Hospital , Istanbul , Turkey
| | - Burak Tatlı
- a Department of Pediatric Neurology , Istanbul University , Istanbul , Turkey
| |
Collapse
|
45
|
The Molecular Pathway of Argon-Mediated Neuroprotection. Int J Mol Sci 2016; 17:ijms17111816. [PMID: 27809248 PMCID: PMC5133817 DOI: 10.3390/ijms17111816] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/17/2016] [Accepted: 10/25/2016] [Indexed: 11/28/2022] Open
Abstract
The noble gas argon has attracted increasing attention in recent years, especially because of its neuroprotective properties. In a variety of models, ranging from oxygen-glucose deprivation in cell culture to complex models of mid-cerebral artery occlusion, subarachnoid hemorrhage or retinal ischemia-reperfusion injury in animals, argon administration after individual injury demonstrated favorable effects, particularly increased cell survival and even improved neuronal function. As an inert molecule, argon did not show signs of adverse effects in the in vitro and in vivo model used, while being comparably cheap and easy to apply. However, the molecular mechanism by which argon is able to exert its protective and beneficial characteristics remains unclear. Although there are many pieces missing to complete the signaling pathway throughout the cell, it is the aim of this review to summarize the known parts of the molecular pathways and to combine them to provide a clear insight into the cellular pathway, starting with the receptors that may be involved in mediating argons effects and ending with the translational response.
Collapse
|
46
|
|
47
|
Fahlenkamp AV, Rossaint R, Coburn M. [Neuroprotection by noble gases: New developments and insights]. Anaesthesist 2016; 64:855-8. [PMID: 26329914 DOI: 10.1007/s00101-015-0079-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Noble gases are chemically inert elements, some of which exert biological activity. Experimental neuroprotection in particular has been widely shown for xenon, argon and helium. The underlying mechanisms of action are not yet fully understood. Besides an interference with neuronal ion-gated channels and cellular signaling pathways as well as anti-apoptotic effects, the modulation of neuroinflammation seems to play a crucial role. This review presents the current knowledge on neuroprotection by noble gases with a focus on interactions with the neuronal-glial network and neuroinflammation and the perspectives on clinical applications.
Collapse
Affiliation(s)
- A V Fahlenkamp
- Klinik für Anästhesiologie, Universitätsklinikum Aachen, RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland.
| | - R Rossaint
- Klinik für Anästhesiologie, Universitätsklinikum Aachen, RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland
| | - M Coburn
- Klinik für Anästhesiologie, Universitätsklinikum Aachen, RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland
| |
Collapse
|
48
|
Hafner C, Qi H, Soto-Gonzalez L, Doerr K, Ullrich R, Tretter EV, Markstaller K, Klein KU. Argon Preconditioning Protects Airway Epithelial Cells against Hydrogen Peroxide-Induced Oxidative Stress. Eur Surg Res 2016; 57:252-262. [PMID: 27560977 DOI: 10.1159/000448682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 07/25/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Oxidative stress is the predominant pathogenic mechanism of ischaemia-reperfusion (IR) injury. The noble gas argon has been shown to alleviate oxidative stress-related myocardial and cerebral injury. The risk of lung IR injury is increased in some major surgeries, reducing clinical outcome. However, no study has examined the lung-protective efficacy of argon preconditioning. The present study investigated the protective effects of argon preconditioning on airway epithelial cells exposed to hydrogen peroxide (H2O2) to induce oxidative stress. METHODS A549 airway epithelial cells were treated with a cytotoxic concentration of H2O2 after exposure to standard air or 30 or 50% argon/21% oxygen/5% carbon dioxide/rest nitrogen for 30, 45 or 180 min. Cells were stained with annexin V/propidium iodide, and apoptosis was evaluated by fluorescence-activated cell sorting. Protective signalling pathways activated by argon exposure were identified by Western blot analysis for phosphorylated candidate molecules of the mitogen-activated protein kinase and protein kinase B (Akt) pathways. RESULTS Preconditioning with 50% argon for 30, 45 and 180 min and 30% argon for 180 min caused significant protection of A549 cells against H2O2-induced apoptosis, with increases in cellular viability of 5-47% (p < 0.0001). A small adverse effect was also observed, which presented as a 12-15% increase in cellular necrosis in argon-treated groups. Argon exposure resulted in early activation of c-Jun N-terminal kinase (JNK) and p38, peaking 10- 30 min after the start of preconditioning, and delayed activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, peaking after 60-90 min. CONCLUSIONS Argon preconditioning protects airway epithelial cells from H2O2-induced apoptotic cell death. Argon activates the JNK, p38, and ERK1/2 pathways, but not the Akt pathway. The cytoprotective properties of argon suggest possible prophylactic applications in surgery-related IR injury of the lungs.
Collapse
Affiliation(s)
- Christina Hafner
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Heme Oxygenase-1 Mediates Neuroprotection Conferred by Argon in Combination with Hypothermia in Neonatal Hypoxia–Ischemia Brain Injury. Anesthesiology 2016; 125:180-92. [DOI: 10.1097/aln.0000000000001128] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
Background
Hypoxic–ischemic encephalopathy is a major cause of mortality and disability in the newborn. The authors investigated the protective effects of argon combined with hypothermia on neonatal rat hypoxic–ischemic brain injury.
Methods
In in vitro studies, rat cortical neuronal cell cultures were challenged by oxygen and glucose deprivation for 90 min and exposed to 70% Ar or N2 with 5% CO2 balanced with O2, at 33°C for 2 h. Neuronal phospho-Akt, heme oxygenase-1 and phospho-glycogen synthase kinase-3β expression, and cell death were assessed. In in vivo studies, neonatal rats were subjected to unilateral common carotid artery ligation followed by hypoxia (8% O2 balanced with N2 and CO2) for 90 min. They were exposed to 70% Ar or N2 balanced with oxygen at 33°, 35°, and 37°C for 2 h. Brain injury was assessed at 24 h or 4 weeks after treatment.
Results
In in vitro studies, argon–hypothermia treatment increased phospho-Akt and heme oxygenase-1 expression and significantly reduced the phospho-glycogen synthase kinase-3β Tyr-216 expression, cytochrome C release, and cell death in oxygen–glucose deprivation–exposed cortical neurons. In in vivo studies, argon–hypothermia treatment decreased hypoxia/ischemia-induced brain infarct size (n = 10) and both caspase-3 and nuclear factor-κB activation in the cortex and hippocampus. It also reduced hippocampal astrocyte activation and proliferation. Inhibition of phosphoinositide-3-kinase (PI3K)/Akt pathway through LY294002 attenuated cerebral protection conferred by argon–hypothermia treatment (n = 8).
Conclusion
Argon combined with hypothermia provides neuroprotection against cerebral hypoxia–ischemia damage in neonatal rats, which could serve as a new therapeutic strategy against hypoxic–ischemic encephalopathy.
Collapse
|
50
|
|