Next Article in Journal
Recombinase Polymerase Amplification (RPA) of CaMV-35S Promoter and nos Terminator for Rapid Detection of Genetically Modified Crops
Next Article in Special Issue
Investigation of the Neuroprotective Impact of Nimodipine on Neuro2a Cells by Means of a Surgery-Like Stress Model
Previous Article in Journal
Towards Global QSAR Model Building for Acute Toxicity: Munro Database Case Study
Previous Article in Special Issue
Sildenafil Attenuates Inflammation and Oxidative Stress in Pelvic Ganglia Neurons after Bilateral Cavernosal Nerve Damage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Argon: Systematic Review on Neuro- and Organoprotective Properties of an “Inert” Gas

by
Anke Höllig
1,2,
Anita Schug
1,
Astrid V. Fahlenkamp
2,
Rolf Rossaint
2,
Mark Coburn
2,* and
Argon Organo-Protective Network (AON)
1
Department of Neurosurgery, University RWTH Aachen, 52074 Aachen, Germany
2
Department of Anesthesiology, University RWTH Aachen, 52074 Aachen, Germany
*
Author to whom correspondence should be addressed.
Members are listed in Appendix.
Int. J. Mol. Sci. 2014, 15(10), 18175-18196; https://doi.org/10.3390/ijms151018175
Submission received: 14 August 2014 / Revised: 12 September 2014 / Accepted: 23 September 2014 / Published: 10 October 2014
(This article belongs to the Special Issue Neuroprotective Strategies 2014)

Abstract

:
Argon belongs to the group of noble gases, which are regarded as chemically inert. Astonishingly some of these gases exert biological properties and during the last decades more and more reports demonstrated neuroprotective and organoprotective effects. Recent studies predominately use in vivo or in vitro models for ischemic pathologies to investigate the effect of argon treatment. Promising data has been published concerning pathologies like cerebral ischemia, traumatic brain injury and hypoxic ischemic encephalopathy. However, models applied and administration of the therapeutic gas vary. Here we provide a systematic review to summarize the available data on argon’s neuro- and organoprotective effects and discuss its possible mechanism of action. We aim to provide a summary to allow further studies with a more homogeneous setting to investigate possible clinical applications of argon.

1. Introduction

Argon belongs to the noble gases and generally is regarded as an inert, non-reactive element. Even its name (from the Greek “αργός”—inert) refers to its chemical inactivity. In fact, biological effects of the noble gases including argon have been identified starting in the 1930s: its narcotic properties under hyperbaric circumstances were described beginning with studies investigating argon as a possible breathing gas for divers [1]. Recently, neuroprotective and organoprotective features have been identified [2,3,4,5,6].
In general, most promising therapeutics—especially neuroprotectants—identified through preclinical studies have failed to demonstrate efficacy in clinical trials due to heterogeneous experimental settings, inadequate sample sizes, inappropriate time and dosage of application and so on [7,8]. Concerning argon’s beneficial properties, most of the evidence has been accomplished by in vitro, in vivo and rarely human studies. Again, the multitude of anecdotal reports and experimental models applied hinders the overall assessment of argon’s therapeutic potential but also its possible side effects. Therefore we performed a systematic review on the current literature on argon. We provide an overview of available data on argon’s organoprotective and particularly its neuroprotective features as well as potential side effects. Further, we illustrate the current data on the possible mechanism of action and future perspectives for therapeutic applications of argon.

2. Results

The PubMed search revealed 671 hits, from which 42 records were identified as relevant for screening. The alternative databases (Embase, Scisearch, Biosys, gms) presented 1501 records using the same search strategy. Eighty-seven records were regarded relevant. Thirty-five articles had to be excluded with regard to content (review articles, comments or articles on technical applications of argon, abstracts and poster presentations); one article had to be rejected as only available in the Chinese language. Duplicates (n = 65) among the two database searches were eliminated. In Figure 1 the procedure is summarized. In total, 38 relevant full text articles were identified. Eleven out of 38 (29%) studies were conducted before, and 27 (71%) after the year 2000. Human studies are scarce (n = 6, see Table 1) and most of them had been motivated by technical considerations in the context of diving or aerospace. In vivo animal experiments dealing with the effects of argon are much more common (n = 22, summarized in Table 2) and the number of publications on in vivo data has increased recently (16 out of 22 articles have been published later than 2000). Most animal experiments were carried out with rats (16 out of 22); in two studies, Japanese quail eggs were used. In vitro studies are dominated by the use of murine organotypic brain slices (4 out of 10 studies; see Table 3).
Figure 1. Diagram showing literature search procedure and results.
Figure 1. Diagram showing literature search procedure and results.
Ijms 15 18175 g001
Comparisons with other noble gases were drawn in 7 (out of 22) animal studies and 5 in vitro studies. Frequently the effect of argon was compared to that of helium (n = 7) and xenon (n = 5).
In human studies, descriptions of argon’s narcotic effect and the possible increase of resistance against hypoxia were most common, whereas among in vivo animal studies, the neuroprotective or organoprotective properties of argon were the main topic (11 out of 22). In general most of the studies dealt with argon’s narcotic effect and the reaction of organisms to hypoxia under argon atmosphere (n = 14). Notably most of these studies were carried out before the year 2000 (9 out of 14). Neuroprotection and organoprotection are relatively new topics: All of the studies covering these topics (n = 17) were carried out after the year 2000. Neuroprotection is, with 11 articles, the field of interest most frequently highlighted in the recent years. Besides tissue protection, recent studies often dealt with the identification of argon’s mechanism of action. In total, 11 investigations addressed this question with 10 of them carried out after 2000. Most studies concerning protective effects of argon and its mechanism of action were carried out using animal or in vitro models.
Notably, argon failed to show protective properties in two studies [9,10], whereas other studies on tissue protection could only demonstrate a partial benefit of argon treatment (i.e., only functional improvement or benefit under certain circumstances like timing of applications) [11,12].

3. Discussion

Our systematic review has highlighted various studies on argon’s effects applying heterogeneous models and questions. We will discuss similarities and differences of the approaches and results.

3.1. Physiological Studies

The first descriptions of argon’s biological effects arose in the context of diving medicine. Mental impairment at high pressures had been observed. Behnke and Yarbrough in 1939 tried to elucidate the role of argon in producing narcotic effects in humans [1]. The first physiological data (like respiratory resistance) could be assessed. Further studies were carried out evaluating mental performance and subjective rating of condition as measurement of narcosis with 80% argon and 20% oxygen under different pressure levels [13]. In humans, long-term effects (up to 7 days) under hyperbaric argon atmosphere were examined demonstrating improved work performance and a shift in lipid metabolism. An increased resistance to hypoxic hypoxia under argon atmosphere was suggested [14]. These results were confirmed by a study testing human oxygen consumption during physical exercise breathing a gas mix with 30% argon. An increase of oxygen consumption under argon was observed, therefore a catalytic activity of argon on oxygen kinetics was supposed [15]. Another long-term study carried out for 9 days (14% oxygen, 33% nitrogen, 54% argon and 0.2% carbon dioxide for 6 days followed by 10% oxygen, 35% nitrogen, 55% argon and 0.2% carbon dioxide) demonstrated no detrimental influence on work performance [16].
Narcotic potency was also examined in mice [9] and rats [17,18]. Therefore metabolism, oxygen consumption and resistance towards hypoxia under argon (different species and organ slices) were investigated [9,19,20,21]. A favorable metabolic condition with a distinct energy metabolism and elevated oxygen consumption was supposed, thus resulting in increased resistance towards hypoxia [20,22]. Furthermore, an improved survival of animals under hypoxic atmosphere was demonstrated [23,20], whereas an earlier series of experiments with white mice did not indicate a beneficial effect of argon on survival [9]. Under hypoxic atmosphere containing argon a change in development (faster development and less teratogenic pathologies) was observed, which also has been attributed to the change of metabolism under argon [19,24,25].
In conclusion, argon seems to improve resistance towards hypoxia. Metabolic changes, cell membrane dependent mechanisms, recovery of mitochondrial enzymes and oxygen synergism have been discussed to explain this phenomenon. Studies concerning mechanism of action will be discussed hereafter in more detail.
Table 1. Human studies.
Table 1. Human studies.
Experimental ModelNumber of CasesDose and ConcentrationOutcome ParameterResults of ExperimentsConclusionReference
Mental performance in Ar-N2-O2-atmospheren = 4, malesix days (5 m depths): 14% O2, 33% N2, 54% Ar, 0.2% CO2 followed by three days: 10% O2, 35% N2, 55% Ar, 0.2% CO2Adaptive biocontrol of cortical (ABC) bioelectric activity synchronization, emotional and mental performance (Luscher test), “Minesweeper” and “Tetris” performancePartial improvement of performance, overall no decrease of ABC skillDespite fluctuations of anxiety levels no influence on work performance, tendency to loose preservation of adaptation process with argon-mixAntonov & Ershova (2009) [16]
Assessment of mental impairment breathing argon at different pressures (corresponding to 90–130 m diving depth)n = 469% Ar, 11% N2, 20% O2, duration not specifiedSelf-assessment of diving depthNo effect on mental status for normobaric argon, mental impairment at pressure levels corresponding to depths of 90–130 m (tendency to overestimate diving depth)Narcotic effect of argon is greater than that of nitrogenBehnke & Yarbrough (1939) [1]
Comparison of argon and nitrogen narcosis at 1 to 10 ATA (0.1 resp. 1.1 MPa)n = 1080% Ar, 20% O2 or air (different pressure levels)Assessment of narcosis: mental arithmetic, subjective estimate of narcosis, adjective checklist.Arithmetic: numbers of errors increase with high pressure (with argon mix more than with air), subjective rating of narcosis: increases with higher pressure (with argon mix more than with air), adjective checklist: number of responses increases with pressure (highly variable)Inert gases exert qualitavely identical effectsFowler & Ackles (1972) [13]
(a) Exposition to white noise (85 dB) for 1 h;
(b) Exposition of rats to hypoxic gas mix;
(c) Exposition of hair cells (ex vivo) to hypoxic Ar-/N2-saturated medium
n = 10(a) 24% Ar, 60% N2, 16% O2, normobaric, duration not specified;
(b) ≥25% Ar, 4%–5% O2 normobaric;
(c) 95% Ar, 5% CO2 or 95% N2, 5% CO2
(a) Pure-tone audiometry, TEOAE, DPOAE, BERA, EcohG;
(b) Survivability of rats;
(c) Survival time of hair cells in medium
(a) Improved condition of acoustic system in the argon treated group;
(b) Increased survival in Ar-gas mix;
(c) Increased survival of hair cells in Ar-containing medium
Oto- and neuroprotective effect of argon, attenuates effects of hypoxiaMatsnev et al. (2007) [23]
Long term (7 day) effects of hypoxic argon-oxygen mixture on human performancen = 4 male7 days (10 m depths): 0.2 kg/cm2 O2, 0.8 kg/cm2 N2, 1.0 kg/cm2 ArAssessment of respiratory, cardiovascular and neurological parameters, evaluation of physical and mental work performanceShift in lipid metabolism, better work performance with hyperbaric 15% Ar-O2 mixtureArgon is physiologically active causing increased resistance to hypoxic hypoxia (redox-reaction)Pavlov et al. (1999) [14]
Oxygen consumption breathing Ar-containing gas mixtures during physical (submaximal) exercisen = 7, male15% O2, 30% Ar, 55% N2 or 15% O2, 85% N2Oxygen consumption, heart rate, ventilation frequency during physical exercise breathing hypoxic gas mixturesIncrease of oxygen consumption during exercise breathing Ar-mix compared to N2-mixCatalytic activity of argon on kinetics of oxygen consumption which might increase tolerance towards hypoxiaShulagin et al. (2001) [15]
Ar = argon; O2 = oxygen; N2 = nitrogen; CO2 = carbon dioxide; TEOAE = transitory evoked otoacoustic emission; DPOAE = distortion product otoacoustic emissions; BERA = brainstem evoked response audiometry; EcohG = electrocochleography.
Table 2. Animal experiments.
Table 2. Animal experiments.
Experimental ModelSpecies, AgeNumber of CasesPressure, Dose and ConcentrationOutcome ParameterResults of ExperimentsConclusionReference
Assessment of argon’s narcotic potency after pretreatment with GABA-antagonists (GABAA-receptor-antagonist gabazine; GABAB-receptor antagonist, GABAA-receptor antagonist benzodiazepine site)Sprague Dawley rats, adultn = 6 per groupArgon was dosed at 0.1 Mpa/min until narcosis was reachedLoss of righting reflexIncrease of argon threshold pressure after pretreatment with GABAA-receptor antagonist and GABAA-receptor antagonist for benzodiazepine siteArgon may interact directly with the GABAA receptor and partly with its benzodiazepine siteAbraini et al. (2003) [26]
Evaluation of relationship of locomotor and motor activity and striatal dopamine release under argon narcosisSprague Dawley rats, adultTotal n = 1082 MPa (with 0.1 MPa/min)Behavioral analysis, quantification of striatal dopamine releaseBiphasic pattern with initial hyperactivity after compression; decrease of activity and dopamine release after 1 MPaDopamine release could be related to decrease of hyperactivity under argon narcosisBalon et al. (2003) [27]
Assessment of reaction in response to minimal electroshock and antagonisation with antipsychotic drug (Frenquel)Wistar ratsn = 46; 102 experiments12.6 atm abs (=1.3 MPa)Reaction to minimal electroshockGreater narcotic potency of argon compared with nitrogen, partly abolished by FrenquelArgon narcosis may arise from histotoxic hypoxia; Frenquel somehow decreases the narcotic effectBennett (1963) [17]
Cardiac arrest for 7 min followed by 3 min resuscitation (CPR), postconditioning with argonSprague Dawley rats, adultn = 7 per group1 h after CPR: 70% Ar, 30% O2 for 1 hNeurological performance 7d after CPR, hippocampal cell lossBetter neurological performance (NDS score) and less neuronal damage of neocortex and hippocampus (C3/4), no difference in caspase 3/9 expressionLong lasting functional effect paralleled by less neuronal damage C3/4Brücken et al. (2013) [28]
Cardiac arrest for 7 min followed by 3 min resuscitation (CPR), postconditioning with argon, pretreatment with 5HD (KATP-Channel-Blocker)Sprague Dawley rats, adultn = 9 per group1 h after CPR: either 70% Ar and 30% O2 or 40% Ar, 30% O2 and 30% N2Neurological performance 8d after CPR, neuronal loss (neocortex, hippocampal C3/4)Better neurological performance in argon –treated group (70% Ar > 40% Ar), less neuronal loss (regardless of Ar-concentration), no influence of 5HD on beneficial argon effectArgon exerts dose dependent neuroprotective effect, KATP-Channels seem not to be involved in the mechanism of actionBrücken et al. (2014) [29]
Cardiac arrest for 7 min followed by 3 min resuscitation (CPR), postconditioning with argonSprague Dawley rats, adultn = 8 per group1h of 70% Ar and 30% O2 either 1 or 3 h after CPR or no argon treatmentNeurological performance 8d after CPR, neuronal loss (neocortex, hippocampal C3/4, basal ganglia)Better neurological performance and less neuronal loss in neocortex and hippocamplas C3/4 in both argon—treated groups, less neuronal damage in basal ganglia (3 h delay)Argon exerts a neuroprotective effect even after treatment delayed for 3 hBrücken et al. (2014) [30]
Assessment of oxygen consumption and development time of different speciesYeast, Drosophila, Mouse, Zootermopsis, Tenebrio, Cnemidophorus, Coloenyx 80% Ar, 20% O2Oxygen consumption of different species, development time of larvaeArgon alters rate of metabolism and development (acceleration of metamorphosis) in some animalsArgon–either at atmospheric or high pressure is not inertCook (1950) [19]
(a) OGD (brain slices);
(b) NMDA-induced brain damage (in vivo);
(c) MCAO (in vivo)
Sprague Dawley rats, adultn = 8 to n= 14 per group(a) 15%–75% Ar for 3 h after OGD;
(b) 15%–75% Ar for 3 h (1 h after NMDA);
(c) 50% Ar, 25% N2, 25% O2 for 3 h (2 h after MCAO)
(a) LDH release after OGD;
(b) Extent of brain damage;
(c) Neurologic outcome and extent of brain damage
(a) Most pronounced reduction of LDH release compared to N2 in 50% argon treated (less with 37.5% and 75% Ar);
(b) Significantly attenuation of NMDA induced brain damage with 37.5 and 50% Ar;
(c) Reduction of cortical ischemic volume by Ar, increase of subcortical brain damage, decrease of neurological score compared to sham
Argon shows antiexcitotoxic effects (oxygen like properties), but due the demonstrated adverse effects (increase of subcortical damage. and decrease of neurological function in the argon treated group after MCAO) results do not support therapeutic postischemic application of argon, protective effect after NMDA-induced brain injury and OGD.David et al. (2012) [11]
2 h of MCAO, 1 h after MCAO either 50% Ar/50% O2 or 50% N2/50% O2Sprague-Dawley rats, adultn = 5350% Ar/50% O2 or 50% N2/50% O2 for 1 h, normobaric24 h after MCAO, expression analysis of inflammatory and growth factors, cell count of neurons, astrocytes and microgliaIn argon-treated MCAO significantly higher expression levels of IL-1beta, IL-6, iNOS, TGF-beta, and NGF were found compared to MCAO. VEGF was significantly elevated compared to sham. Significant reduction of neurons only occurred in the penumbra after MCAOAn elevated expression of several inflammatory and growth factors following MCAO + argon compared to MCAO + placebo and shamFahlenkamp et al. (2014) [31]
Effect of hypoxic argon containing gas mix (for 4 days) on early embryogenic developmentJapanese quail eggsn = 3015% O2, 30% N2, 55% Ar or 15% O2, 85% N2 for 4 daysAssessment of survival and developmentWith argon containing gas mix up to 60% development, normal morphology, without argon only 17% reached adequate developmental state Positive effect of argon on embryonic development in hypoxic atmosphereGur’eva et al. (2008) [24]
Transplantation of harvested kidneys after storage in Ar-, Xe- or N2-saturated solutionWistar rats, adultn = 60Storage in Ar-, Xe- or N2-saturated solution for 6 hAssessment of renal function (Creatinine clearance, urinary albumin) 7 and 14 days after transplantation, histological examination of transplanted kidneys 14 days after transplantationCreatinine clearance higher and urinary albumin lower as well as better renal architecture in Ar-treated group compared to N2 treated with a more pronounced effect by argon than by xenon treatmentDecrease of ischemia-reperfusion injury, improved graft function and maintained anatomical structure after Ar- treatment (compared to Xe and N2)Irani et al. (2011) [32]
LAD occlusion for 30 min, preconditioning with 70% Ar/He/Ne/30% O2 or hypoxic preconditioningNew Zealand white, rabbitn = 98Preconditioning with 3 cycles each 5 min (70% Ar/He/Ne, 30% O2), normobaricAssessment of infarct size compared to hypoxic preconditioning compared to control (no preconditioning)Significant reduction of infarct size after preconditioning with Ar, He and NeMore pronounced cardioprotection with Ar-preconditioning compared to hypoxic preconditioningPagel et al. (2007) [33]
LAD occlusion, cardiac arrest for 8 min, CPR for 5 min followed by defibrillation, postconditioning for 4 h with either Ar/O2 or N2/O2.Domestic pig, malen = 1270% Ar, 30% O2 or 70% N2, 30% O2 for 4 h, normobaricAssessment of survival and neurological function 72 h after CPR, serum neuron-specific enolase (NSE) and troponin, Immunohistochemistry of brain slicesBetter neurological performance in argon-treated group, significantly lower increase in serum NSE and minimal histological brain injuryFaster, complete neurologic recovery with argon treatment, no detrimental side effects, mainly functional improvement assessedRistagno et al. (2014) [12]
Narcotic effect of compression in argon atmosphereRats, 15 weeksn = 15Ar 100–800 kPaAssessment of behavior during compression and decompressionFirst signs of narcosis from 500 kPa on, subsequently falling asleep at 800 kPa (8 of 10 animals)Demonstration of narcotic properties of argonRužička et al. (2007) [18]
2 h of MCAO, 1 h after MCAO either 50% Ar/50% O2 or 50% N2/50% O2Sprague Dawley rats, adultn = 2250% Ar/50% O2 or 50% N2/50% O2 for 1 h, normobaric24 h after MCAO: neurological assessment, evaluation of infarct sizeImproved composite adverse outcome, reduction of infarct volume (overall, cortical and subcortical) in argon-treated groupArgon demonstrates in vivo neuroprotective properties (reduced infarct size), but no improvement concerning neurological outcome and mortalityRyang et al. (2011) [34]
Survivability of rats in hypoxic argon containing atmosphereWistar rats Hypoxic atmosphere: O2 (4%–8%), different concentrations of Ar (0%–80%), N2 (15%–87%) and CO2 (0%–8%)Survival rate of rats in hypoxic atmospheres with different gas mixAdding argon increases survival rate, adding CO2 and increasing temperature reduces survival rateAdding argon improves hypoxic toleranceSoldatov et al. (1998) [20]
Effect of hypoxic environment on developmentJapanese quail eggs 10% O2, 55% Ar, 35% N2 or 10% O2, 90% N2Assessment of survival rate and occurrence of teratogenic pathologiesArgon containing gas mixture reduces occurrence of teratogenic events, 100% mortality after 7 days with both mixturesArgon reduces incidence of teratogenic events probably by stimulation of metabolismSoldatov et al. (2002) [25]
Influence of hypoxic atmosphere (O2/Ar or O2/N2) on brain metabolismWhite rats Hypoxic atmosphere: O2 (7%) with Ar or N2Detection of NADH/NAD in brain slicesArgon attenuates hypoxia induced metabolic impairmentPositive effect on cerebral energy metabolism by argonVdovin et al. (1998) [21]
Decompression in atmospheres containing Ar or HeMale albino miceTotal n= 23179% Ar/ He, 21% O2, decompression to 179 mmHgSurvival rate during decompression at different temperatures, assessment of oxygen consumptionSurvival rate in argon containing atmosphere similar to air during decompression, higher survival rate in helium containing atmosphereHelium promotes hypoxic resistance of mice, but none observed for argonWitherspoon et al. (1964) [9]
Hypoxic ischemic brain injury: ligation of right carotid artery, hypoxia (8% O2, 92% N2) 1h after ligation for 90 min (moderate) or 120 min (severe) followed by postconditioning with Ar/He/Xe or controlSprague Dawley rats, age: 7 daysn = 5 per group120 min after hypoxia: 70% Ar, 30% O2 for 90 min, normobaricCell viability after moderate and severe hypoxia (7 and 14 days thereafter), infarct volume, neurologic/motor performance, protein analysis contralateral hemisphereImproved cell viability with postconditiong (Ar > Xe, He) after moderate hypoxia, improvement after severe hypoxia by Ar and Xe, induction of Bcl-2 (contralateral hemisphere) after Ar-postconditioning, neurologic function in noble gas treated animals better than controlPronounced neuroprotective effect by argon after mild and severe hypoxia, possibly acts via upregulation of Bcl-2 expressionZhuang et al. (2012) [35]
Ar = argon; N2 = nitrogen; O2 = oxygen; Xe = xenon; He = helium; Ne = neon; CO2 = carbon dioxide; GABAA-receptor = gamma-aminobutyric acid A receptor; GABAB-receptor = gamma-aminobutyric acid B receptor; Frenquel = γ-pipradol or Azacyclonol; CPR = cardiopulmonary resuscitation; MCAO = middle cerebral artery occlusion; KATP-Channel = ATP-sensitive potassium channel; OGD = oxygen glucose deprivation; NMDA-receptor = N-Methyl-d-aspartic acid-receptor; MCAO = middle cerebral artery occlusion; LDH = Lactate dehydrogenase; LAD = left anterior descending artery; Bcl-2 = B-cell lymphoma 2.
Table 3. In vitro studies.
Table 3. In vitro studies.
Experimental ModelStudied MaterialPressure, Dose and ConcentrationOutcome ParameterResults of ExperimentsConclusionReference
Evaluation of interaction of argon and tPA (tissue plasminogen activator) on enzymatic and thrombolytic efficiency: catalytic efficiency of tPA, blood clot formation and thrombolysisWhole blood (Sprague Dawley rats)25%–75% Ar, 25% O2Catalytic and thrombolytic efficiency of tPAConcentration dependent dual effect of argon on tPA effect: at concentrations higher than 50% argon increases catalytic and thrombolytic efficiency, but decreases them at concentrations lower 50%Effect may be due to elastase binding of argon or to its interaction with oxygen competing for tPA binding and overcoming the hypoxic effect with higher concentrations (oxygen synergism)David et al. (2013) [36]
Nitrogen or argon hypoxia (OGD) for 90 min followed by postconditioning with argon or nitrogen (each 75%)Foetal (18 days) BALB/c mice, brain slicesOGD: 75% Ar, 20% N2 or 95% N2, 5% CO2; followed by: 75% Ar or 75% N2, 20% O2, 5% CO2, normobaricCell viability quantified by MTT assayNeuroprotective effect of argon after OGD (less than Xe, also tested), in the absence of OGD: improved cell viability with argon compared to control (naïve)Argon shows a significant neuroprotective effect but less pronounced than with xenonJawad et al. (2009) [37]
Exposure of primary neuronal and astroglial cell cultures and the microglial cell line BV-2 to 50% argon, additionally stimulation of microglia with LPSBALB/c mice (primary cultures), BV-2 cell lineExposure of primary cultures to 50% Ar for 15–120 min (vs. control N2 instead of Ar)Protein analysis after treatment and stimulation with LPS, analysis of RNA-expressionIncrease of ERK 1/2 phosphorylation in microglia by argon (mediated by upstream kinase MEK1/2), no phosphotyrosine phosphatase inhibition, no augmentation of LPS-mediated ERK 1/2 activation, no relevant modification of LPS-induced cytokine expression by argonShort enhanced activation of ERK1/2 via MEK by argon (in primary cultures and microglia), activation does not take part via interference with phosphotyrosine phosphatases. No substantial modification of cytokine expression after LPS-exposure in microgliaFahlenkamp et al. (2012) [38]
Membrane stability of peritoneal macrophages (mice) under argon or nitrogen saturated medium after UV-induced damagePeritoneal macrophages (mice)Normobaric, hypoxic Ar or N2 saturated mediumMeasurement of intracellular pH, ability to build up fluoresceinNormobaric environment with Ar or N2 protects plasmatic membranes from UV-induced damageResistance against UV-induced damage is elevated by hypoxic Ar or N2 containing environmentGalchuk et al. (2001) [39]
(a) In vitro traumatic brain injury (hippocampal brain slices), effect of glycine administration; postconditioning with noble gas;
(b) Patch clamp study to evaluate receptor effect.
(a) C57BL/6 mice (brain slices);
(b) HEK293 cells
Different concentrations(a) Extent of cell injury after trauma;
(b) Effect on NMDA-mediated or TREK-1 currents
(a) Argon at 50% atm shows neuroprotective effects attenuate secondary injury after trauma (but less than xenon), glycine does not reverse argon’s positive effect;
(b) NMDA-mediated or Trek-1 currents are not influenced by argon
Argon’s neuroprotective effect seems not to be mediated by NMDA-receptor glycine site, potassium channels neither seem to be involvedHarris et al. (2013) [40]
(a) In vitro trauma (hippocampal brain slice);
(b) OGD for 30 min followed by postconditioning with argon.
Brain slices C57BL/6Postconditioning with 25%-74% argon (directly after lesion) or with 50% argon up to 3 h delayedExtent of cell injury 72 h after lesion(a) Neuroprotective effect of argon after TBI even if applied with delay up to 3 h (most effective at 50% argon concentration);
(b) Dose dependent neuroprotective effect of argon after OGD even if applied with delay up to 3 h
Neuroprotective effect of argon in two types of brain lesions, effect even noticeable with 50% argon after delayed applicationLoetscher et al. (2009) [41]
Oxygen consumption of yeast and liver slices (rat) in inert gas mixtureYeast, liver slices (Sprague Dawley rats)20%–80% ArOxygen consumptionReduced oxygen consumption of yeast and live slices in buffer bubbled with argon, no effect on homogenized liver slicesDepression of oxygen consumption under argon may be due to cell membrane mediated effect as not noticeable with homogenized samplesMaio et al. (1967) [42]
Preconditioning with noble gas (75% for 3 h), 24 h thereafter OGD for 3 hCultured human renal tubular cells (HEK2)75% argon, helium, neon, krypton or xenon for 3 h (24 h after injury)Cell viability 24 h after OGD, without OGD: protein analysis for p-Akt, HIF-1α and Bcl-2No protection from injury by argon, decrease of HIF-1α with argonNo protective effect with argon (but with xenon), for argon: no influence on Bcl-2 expression and decrease of HIF-1α expressionRizvi et al. (2010) [10]
Effect of gas mixtures on induction of apoptotic cell death (by tyrosine kinase inhibitors, DNA-damaging agents and mitochondrial toxins)Human osteosarcoma cells (U2OS)75% Ar or Xe or He or Ne or Kr or N2, 20% O2, 5% CO2Automated fluorescence microscopy to reveal cell deathArgon (and xenon) prevent cell loss after damaging agents, activation of signal transduction pathway sensitive to Z-VAD-fmk, suppresses pathways of intrinsic apoptosis (cytochrome C, caspase 3)Argon suppresses multiple manifestations of the intrinsic apoptotic pathwaySpaggiari et al. (2013) [43]
Trauma of organotypic cultures (organ of Corti, rat) with (a) hypoxia; (b) cisplatin or gentamycinOrganotypic cultures (organ of Corti), Wistar rat(a) 95% Ar or N2, 5% CO2 vs. normoxia;
(b) 74% Ar or N2, 21% O2, 5% CO2
Assessment of cell viability after 48 hLower damage in argon treated group after hypoxia as well as cisplatin or gentamycin damageProtective effect of argon probably affecting Ca+ metabolismYarin et al. (2005) [44]
Ar = argon; N2 = nitrogen; O2 = oxygen; Xe = xenon; He = helium; Ne = neon; CO2 = carbon dioxide; tPA = tissue plasminogen activator; ERK1/2 = extracellular-signal-regulated kinases 1/2; MEK1/2 = MAPKK = mitogen-activated protein kinase kinase; LPS = lipopolysaccharide; NMDA-receptor = N-Methyl-d-aspartic acid-receptor; TREK-1 = Potassium channel subfamily K member 2; p-Akt = phospho-Akt; HIF-1α = hypoxia inducible factor 1α; Bcl-2 = B-cell lymphoma 2; Z-VAD-fmk = pan caspase inhibitor; TBI = traumatic brain injury.

3.2. Neuroprotective and Organoprotective Properties

Within a multitude of experimental models protective effects of argon were investigated: In vitro mostly fetal organotypic murine brain slices were applied. Lesion was induced either by mechanical trauma (in vitro traumatic brain injury-TBI) or by oxygen-glucose-deprivation (OGD) simulating global metabolic stress, i.e., ischemia. Mechanical as well as metabolic stress was diminished by argon application repeatedly [37,40,41,43]. The concentration of argon varied, but averaged at least 50% (in one study 50% atm was applied). Dose dependency for argon treatment after OGD was demonstrated by Loetscher and colleagues, whereas the most effective concentration after in vitro TBI was identified with 50% argon. Even delayed application of postconditioning with argon (up to 3 h after injury) still resulted in decrease of cell death compared to controls without argon treatment [41]. Without injuring the brain slices, application of 75% argon was even able to reduce cell death when compared to controls and showed a less pronounced protective effect than xenon [37]. Another organotypic model assessed hypoxic and toxic resistance of hair cells (organ of Corti) under argon containing atmosphere demonstrating an otoprotective effect [44].
In vivo the most common models are those inducing hypoxia either resulting in cerebral ischemia (by middle cerebral artery occlusion, or hypoxic ischemic brain injury with ligation of carotid artery and exposure to hypoxia) or myocardial ischemia (by LAD-left anterior descending artery-occlusion) or both (by cardiac arrest (CA) followed by delayed resuscitation (CPR)). In line with the clear protective effect after OGD—an in vitro model for cerebral ischemia—Ryang and colleagues [34] demonstrated a reduction of infarct volume and improved composite adverse outcome following argon postconditioning using an MCAO rat model. With the same model (MCAO) but different application time of argon David and colleagues [11] also showed a reduction of cortical infarct volume, but subcortical brain damage increased with argon treatment. In this connection the argon treated animals revealed worse neurological performance (compared to sham), which was found at days 1 and 2 after MCAO. This contrasts with xenon that provides both cortical (greater than argon) and subcortical neuroprotection and further showed improved neurological outcome in the same conditions of MCAO model and timing of treatment [45]. As discussed by David et al., differences between their results and those of Ryang et al. as regards to subcortical neuroprotection could be due to differences in study protocol, particularly timing of treatment (intraischemic vs. postischemic). However, in the same study of David and coworkers, protective effects of argon after OGD were confirmed and, in vivo, an attenuation of NMDA-induced brain damage was shown. Neuroprotective properties after hypoxia (hypoxic ischemic brain injury rat model) were confirmed by Zhuang and colleagues [35]. A more pronounced beneficial effect regarding cell viability for postconditioning with argon was described vis-a-vis nitrogen and even xenon. Combining some features of the aforementioned models, some groups use a resuscitation model to induce cerebral ischemia: In pigs and rats postconditioning with 70% of argon resulted in improved neurological outcome [12,28]. The morphological extent of brain damage (at least for some regions) was reduced compared to controls. In rats dose dependency of the beneficial effect after resuscitation was demonstrated with better neurological outcome after treatment with 70% argon than with 40% [29].
Cardioprotective effects with decrease of infarct size were shown by an in vivo study using argon as a preconditioning drug with a rabbit model [33]. Another possible application of argon is to protect donor organs before transplantation. Rat kidneys harvested in argon saturated solution demonstrated better functional and morphological condition than controls (nitrogen saturated solution) or even xenon treated group [32].
Finally the only human study on neuroprotection was carried out to investigate the effect of argon treatment on exposure to white noise. Improved condition of the acoustic system was shown assuming an oto- and neuroprotective effect of treatment [23]. This hypothesis was strengthened by experimental data on improved hair cell survival with argon treatment.
In conclusion argon’s neuroprotective and organoprotective properties were confirmed by various studies using a multitude of experimental models primarily to simulate hypoxic and less frequently mechanical cell stress. Neuroprotection is the field most commonly covered and most studies underscore the beneficial effect of argon treatment. Nevertheless, results are biased by heterogeneously applied experimental models and differences in study protocols (different timing, concentration and duration of treatment).

3.3. Mechanism of Action

Very little is known about the actual mechanism of action of argon. Abraini and colleagues investigated the involvement of GABA-receptors by examining argon’s narcotic potency in rats after pretreatment with specific GABA-receptor antagonists. They discovered that in a rat model argon threshold pressure had to be increased after pretreatment with GABAA-receptor antagonists and to a lesser extent after GABAA-receptor antagonists for the benzodiazepine site. This was not the case after pretreatment with a GABAB-receptor antagonist. Thus—similar to nitrogen—involvement of GABAA- and the benzodiazepine site of GABAA-receptors, but none of GABAB-receptors, was hypothesized [26]. However, this finding is limited by the fact that Abraini and colleagues used the (hyperbaric) narcotic properties of argon as outcome parameter. Therefore it is problematic transferring the results into the area of neuroprotection, which is achieved under normobaric circumstances. Furthermore at atmospheric pressures, argon did not provoke an intracellular acidosis in macrophages that is induced by other benzodiazepine-sensitive GABAA-receptor agonists [46]. Thus, two distinct, independent methods of action are conceivable dependent on ambient pressure and response measured.
Another in vivo study correlated the extent of striatal dopamine release with the narcotic effect of argon. Decrease of striatal dopamine release was seen in parallel to gas narcosis [27]. Again, this finding relies on argon’s narcotic properties not its cytoprotective properties as indicator of outcome.
Similar to xenon, which inhibits NMDA-receptors [47], this receptor type was investigated during argon treatment. Application of glycine did not reverse the beneficial effect of argon after in vitro TBI, therefore involvement of the glycine site of the NMDA-receptor in argon’s mechanism of action was ruled out. Further, using electrophysiology (patch clamp technique) no effect of argon on NMDA-mediated currents was found, likewise for currents flowing through TREK-1, a two-pore-domain potassium channel [40]. In an in vivo study (resuscitation rat model), pretreatment with a KATP-channel blocker (5-Hydroxydecanoate = 5HD) also failed to impact argon’s beneficial effect [29]. Therefore, neither NMDA receptors nor potassium channels seem to be involved. However, these results will have to be confirmed in further experiments.
Another in vivo study tested in a rat model of hypoxic ischemic brain injury and postconditioning with 70% argon, helium or xenon the expression of three proteins involved in the intrinsic apoptotic pathway: Bax, Bcl-2, and Bcl-xL. Treatment with argon, helium, and xenon increased the expression of Bcl-2. Surprisingly, helium and xenon, with the exception of argon, increased Bcl-xL, a prosurvival protein, whereas expression of Bax, which promotes cell death, was induced after treatment with helium [35]. Again, these results may reflect the uniqueness of each noble gas in regard to its mechanism of action. Further, noble gas modulation of prosurvival proteins has to be elucidated.
Using cultured renal tubular cells (HEK2) prosurvival proteins were investigated in vitro. After preconditioning with 75% helium, neon, argon, krypton and xenon, cell cultures were subjected to OGD. Surprisingly, only xenon treatment showed protection of cell viability. Further, prosurvival proteins (Bcl-2, pAkt -Phospho-Akt- and HIF-1α-hypoxia inducible factor 1 α) were analyzed without OGD. Expression of HIF-1 α increased after treatment with argon, while Bcl-2 and p-Akt expression were not modified. However, xenon treatment led to an increase of all the examined proteins, Bcl-2, p-Akt and HIF-1α [10].
This is in contrast to the results mentioned above and may be due to different experimental settings (in vivo vs. in vitro), different models of tissue stress (hypoxic ischemic brain injury vs. naïve cell culture) and different time points of analysis.
Multiple damaging agents were tested in an in vitro study using a human osteosarcoma cell line (U2OS). Cells were exposed to a tyrosine kinase inhibitor (staurosporine), a DNA-damaging agent (mitoxantrone) and mitochondrial toxins. Argon and xenon inhibited cell loss by staurosporine, mitoxantrone and the mitochondrial toxins, maintained mitochondrial integrity and inhibited caspase-3 expression [43]. Suppression of caspase-3 and cytochrome C once again indicates inhibition of intrinsic apoptotic pathway by argon and xenon.
Using microglial cell cultures and primary neuronal and astroglial cultures the involvement of ERK1/2 (extracellular signal-regulated kinases) with a short and enhanced activation after exposure to 50% argon was demonstrated, but no relevant influence on cytokine expression (contrary to xenon) was found [38].
Finally, protein interactions of argon have to be mentioned: Colloch’h and colleagues investigated the protein-noble gas interactions of xenon, krypton and argon [48]. Three different enzymes were studied showing gas occupancies in the order of their polarizability with highest occupancy reached by xenon and lowest by argon administration, which is similar to the results of Quillin and colleagues examining T4 lysozyme [49]. Depending on the enzyme, different mechanisms of noble gas action were demonstrated: either inhibition of the catalytic reaction through an indirect mechanism, inhibition of the catalytic reaction through a direct mechanism, or prevention of substrate binding. The considerable effects of noble gases are not completely explained by the binding through very weak non-covalent van der Waals interactions. Therefore, the authors conclude that small effects on an array of biological targets may be responsible for the biological effects of noble gases but specific effects (like neuroprotection) of the noble gases may also be due to action via one particular target, which may be specific for each noble gas [48].
In conclusion, argon may distinguish itself from xenon while possibly sharing some joint features during further signaling (like Bcl-2 involvement). Also ERK1/2-signaling plays a role in signal transduction by argon. Decidedly, argon seems not to act via NMDA-receptor signaling or via potassium channels. Although argon would act as a GABAA agonist to induce narcosis as shown in hyperbaric conditions, whether this could apply to normobaric condition as a mechanisms for neuroprotection still remains to be shown. Therefore, the precise target(s) for the biological effects generated by argon administration remains to be elucidated. Only limited evidence indicates the involvement of GABAA-receptor signaling. Finally, approaching the topic from the chemical point of view, one has to highlight the assessment of two important chemists (Nikolai Nikolajewitsch Semjonow and Cyril Norman Hinshelwood), who pointed out the oxygen-like properties of argon: the presence of argon allows reactions between phosphorous and oxygen under pressure levels, which would not happen without argon, therefore acting as sort of catalyst [50]. Thus, increase of resistance towards hypoxia may be explained by argon’s oxygen-like properties as hypothesized by David and colleagues previously [11,36].

3.4. Lack of Clarity

However, while appreciating many promising details of argons possible protective actions, some discrepancies should not be overlooked:
In one in vivo study under hypoxic argon atmosphere, mice did not survive longer than the control group [9]. Another in vitro study using OGD as experimental model did not disclose a beneficial effect of argon preconditioning [10]. Finally, argon treatment in rats applying MCAO resulted in one study in reduced infarct volume (including subcortical area) but in the other in increased infarct volume of subcortical area and worse neurological outcome [11,34]. During one trial the application of argon occurred within the intraischemic phase, and on another occasion after reperfusion, as David and colleagues clearly pointed out [11]. 
These discrepancies may be attributed to differences in the study protocol. One major problem analyzing the studies on argon is that treatment varies between pre-conditioning and post-conditioning. Even if the same “type” of treatment is applied, timing, concentration and duration of administration diverge.
Therefore, to gain more insights into argon’s protective effects as well as identifying its mechanisms of action, standardizing study protocols would be advantageous. Argon’s cytoprotective and special neuroprotective properties have been demonstrated in many studies. Transfer into clinics has not yet occurred due to a lack of data for argon’s practical implementation and potential side effects. David and colleagues [36] tested argon in the context of tPA (tissue-type plasminogen activator) application to review a potential application in stroke therapy. Results demonstrate a dual argon effect. The somehow unexpected inhibiting effect of argon at low concentration on tPA efficiency according to the authors may be due to aforementioned interactions with proteins dependent on multiple factors like gas accessibility and affinity to hydrophobic cavities and the oxygen-like properties of argon [36]. Thus, additionally considering dual effects is necessary for further identification of the appropriate clinical administration concerning timing and duration as well as detection of the mechanism of action.

4. Methods

A PubMed search was carried out in June 2014 with the following search terms: neuroprotection OR organ protection OR cell death OR neuro* OR hypoxic ischemic encephalopathy OR asphyxia OR ischemia OR hypoxia OR ogd OR tbi OR protect* AND argon. Additionally, alternative databases (Embase, Scisearch, Biosys, gms) were screened for the same search terms. Afterwards duplicates were eliminated. The reference lists of review and other relevant articles were hand-searched for appropriate articles and two additional articles, which were later published online ahead of print, were included as well. Of note, Russian articles have been translated by a non-native speaker and therefore we might have caused a translation bias. Additionally, the heterogeneity of experimental settings may hinder the final appraisal.

5. Conclusions

Argon’s neuroprotective and organoprotective properties have been demonstrated repeatedly, but still uncertainties arise from the inhomogeneity of applied models, timing and dosage of argon application.

Acknowledgments

The presented work was supported by the DFG grant CO 799/6-1. We would like to thank Monroe Coburn for language editing the manuscript.

Author Contributions

Anke Höllig conducted database search, summarized the study results and wrote the preliminary draft. Anita Schug translated the Russian articles and helped to elaborate the data available from articles in Russian. Astrid Fahlenkamp and Rolf Rossaint reviewed and revised the manuscript. Mark Coburn revised every single detail of the manuscript and provided overall supervision. The AON group provided a forum to discuss and exchange recent data on argon as therapeutic agent and its mechanism of action.

Appendix

Members of the AON group:
Anne Brücken, University Hospital RWTH Aachen, Aachen, Germany
Michael Fries, University Hospital RWTH Aachen, Aachen, Germany
Oliver Kepp, INSERM, U848, Villejuif, France
Marc Lemaire, Air Liquide Santé, Paris, France
Daqing Ma, Imperial College London, UK
Guy Magalon, Hospital CHU Timone, Marseille, France
Patrick P. Michel, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Epinière, Paris, France
Arne Neyrinck, UZ Leuven, Leuven, Belgium
Jan Pype, Air Liquide Santé, Paris, France
Steffen Rex, UZ Leuven, Leuven, Belgium
Robert D. Sanders, University College London, London, UK
Sinead Savage, Imperial College, London, UK
Christian Stoppe, University RWTH Aachen, Aachen, Gemany

Conflicts of Interest

Conflicts of interest: the aim of the AON meeting was to give an overview on present argon research, to generate a research roadmap and to discuss possible biological mechanisms of argon. The members of the AON group including Anke Höllig and Astrid Fahlenkamp received a refund of travel expenses from Air Liquide Santé International. Mark Coburn and Rolf Rossaint received lecture and consultant fees and refund of travel expenses from Air Liquide Santé International, a company interested in developing clinical applications for medical gases including argon.

References

  1. Behnke, A.R.; Yarbrough, O.D. Respiratory resistance, oil water solubility and mental effects of argon compared with helium and nitrogen. Am. J. Physiol. 1939, 126, 409–415. [Google Scholar]
  2. Nowrangi, D.S.; Tang, J.; Zhang, J.H. Argon gas: A potential neuroprotectant and promising medical therapy. Med. Gas Res. 2014, 4, 3. [Google Scholar] [CrossRef]
  3. Deng, J.; Lei, C.; Chen, Y.; Fang, Z.; Yang, Q.; Zhang, H.; Cai, M.; Shi, L.; Dong, H.; Xiong, L. Neuroprotective gases—Fantasy or reality for clinical use? Prog. Neurobiol. 2014, 115, 210–245. [Google Scholar]
  4. Coburn, M.; Sanders, R.D.; Ma, D.; Fries, M.; Rex, S.; Magalon, G.; Rossaint, R. Argon: The “lazy” noble gas with organoprotective properties. Eur. J. Anaesthesiol. 2012, 29, 549–551. [Google Scholar] [CrossRef] [PubMed]
  5. Coburn, M.; Rossaint, R. Argon in the fast lane: Noble gases and their neuroprotective effects. Crit. Care Med. 2012, 40, 1965–1966. [Google Scholar] [CrossRef] [PubMed]
  6. Pavlov, N.B. Argon—A biologically active component of atmosphere. Aerosp. Environ. Med. 2006, 40, 3–6. [Google Scholar]
  7. Fisher, M. New approaches to neuroprotective drug development. Stroke; J. Cereb. Circ. 2011, 42, S24–S27. [Google Scholar] [CrossRef]
  8. O’Collins, V.E.; Macleod, M.R.; Donnan, G.A.; Horky, L.L.; van der Worp, B.H.; Howells, D.W. 1026 experimental treatments in acute stroke. Ann. Neurol. 2006, 59, 467–477. [Google Scholar] [CrossRef] [PubMed]
  9. Witherspoon, J.D.; Wiebers, J.E.; Hiestand, W.A.; Heimlich, A.H. Decompression of mice in atmospheres containing helium or argon in place of nitrogen. Aerosp. Med. 1964, 35, 529–532. [Google Scholar] [PubMed]
  10. Rizvi, M.; Jawad, N.; Li, Y.; Vizcaychipi, M.P.; Maze, M.; Ma, D. Effect of noble gases on oxygen and glucose deprived injury in human tubular kidney cells. Exp. Biol. Med. 2010, 235, 886–891. [Google Scholar] [CrossRef]
  11. David, H.N.; Haelewyn, B.; Degoulet, M.; Colomb, D.G., Jr.; Risso, J.J.; Abraini, J.H. Ex vivo and in vivo neuroprotection induced by argon when given after an excitotoxic or ischemic insult. PLoS One 2012, 7, e30934. [Google Scholar]
  12. Ristagno, G.; Fumagalli, F.; Russo, I.; Tantillo, S.; Zani, D.D.; Locatelli, V.; de Maglie, M.; Novelli, D.; Staszewsky, L.; Vago, T.; et al. Postresuscitation treatment with argon improves early neurological recovery in a porcine model of cardiac arrest. Shock 2014, 41, 72–78. [Google Scholar] [CrossRef] [PubMed]
  13. Fowler, B.; Ackles, K.N. Narcotic effects in man of breathing 80–20 argon-oxygen and air under hyperbaric conditions. Aerosp. Med. 1972, 43, 1219–1224. [Google Scholar] [PubMed]
  14. Pavlov, B.N.; Buravkov, S.V.; Soldatov, P.E.; Vdovin, A.V.; Deviatova, N.V. The effects of oxygen-argon gaseous mixtures on humans under long-term hyperbaric condition. In Advances in High Pressure Bioscience and Biotechnology; Springer: Berlin Heidelberg, Germany, 1999; pp. 561–564. [Google Scholar]
  15. Shulagin, Iu.A.; D’Iachenko, A.I.; Pavlov, B.N. Effect of argon on oxygen consumption during physical load under hypoxic conditions in humans. Fiziol. Cheloveka 2001, 27, 95–101. [Google Scholar] [PubMed]
  16. Antonov, A.A.; Ershova, T.A. Retention of the skill to perform adaptive bio-control of bioelectrical activity synchronization in the human brain cortex in an argon-nitrogen-oxygen atmosphere with various oxygen content. Aerosp. Environ. Med. 2009, 43, 27–31. [Google Scholar]
  17. Bennett, P.B. Prevention in rats of narcosis produced by inert gases at high pressures. Am. J. Physiol. 1963, 205, 1013–1018. [Google Scholar] [PubMed]
  18. Ruzicka, J.; Benes, J.; Bolek, L.; Markvartova, V. Biological effects of noble gases. Physiol. Res./Acad. Sci. Bohemoslov. 2007, 56, S39–S44. [Google Scholar]
  19. Cook, S.F. The effect of helium and argon on metabolism and metamorphosis. J. Cell. Physiol. 1950, 36, 115–127. [Google Scholar] [CrossRef] [PubMed]
  20. Soldatov, P.E.; D’Iachenko, A.I.; Pavlov, B.N.; Fedotov, A.P.; Chuguev, A.P. Survival of laboratory animals in argon-containing hypoxic gaseous environments. Aerosp. Environ. Med. 1998, 32, 33–37. [Google Scholar]
  21. Vdovin, A.V.; Nozdracheva, L.V.; Pavlov, B.N. Parameters of energy metabolism of the rat brain during inhalation of hypoxic mixtures containing nitrogen and argon. Bull. Exp. Biol. Med. 1998, 125, 618–619. [Google Scholar] [CrossRef]
  22. Pavlov, B.N.; Grigoriev, A.I.; Smolin, V.V.; Komordin, I.P.; Sokolov, G.M.; Ramazanov, R.R.; Spirkov, P.S.; Soldatov, P.E.; Vdovin, A.V.; Buravkova, L.B. Investigations of different hyperoxic, hypoxic and normoxic oxygen-argon gaseous mixtures under different barometric pressure and respiration period. In Proceedings of the 5th International Meeting on High Pressure Biology, St. Petersburg, Russia, 1997; University of Rochester Press: Rochester, NY; USA, 1998; pp. 133–142. [Google Scholar]
  23. Matsnev, E.I.; Sigaleva, E.E.; Tikhonova, G.A.; Buravkova, L.B. Otoprotective effect of argon in exposure to noise. Vestn. Otorinolaringol. 2007, 3, 22–26. [Google Scholar] [PubMed]
  24. Gur’eva, T.S.; Dadasheva, O.A.; Soldatov, P.E.; Sychev, V.N.; Mednikova, E.I.; Smirnov, I.A.; Smolenskaia, T.S.; Dadasheva, M.T. Effect of hypoxic argon-containing gas mixtures on developing organism. Aerosp. Environ. Med. 2008, 42, 40–43. [Google Scholar]
  25. Soldatov, P.E.; Dadasheva, O.A.; Gur’eva, T.S.; Lysenko, L.A.; Remizova, S.E. The effect of argon-containing hypoxic gas environment on development of Japanese quail embryos. Aerosp. Environ. Med. 2002, 36, 25–28. [Google Scholar]
  26. Abraini, J.H.; Kriem, B.; Balon, N.; Rostain, J.C.; Risso, J.J. Gamma-aminobutyric acid neuropharmacological investigations on narcosis produced by nitrogen, argon, or nitrous oxide. Anesth. Analg. 2003, 96, 746–749. [Google Scholar] [CrossRef] [PubMed]
  27. Balon, N.; Risso, J.J.; Blanc, F.; Rostain, J.C.; Weiss, M. Striatal dopamine release and biphasic pattern of locomotor and motor activity under gas narcosis. Life Sci. 2003, 72, 2731–2740. [Google Scholar] [CrossRef] [PubMed]
  28. Brucken, A.; Cizen, A.; Fera, C.; Meinhardt, A.; Weis, J.; Nolte, K.; Rossaint, R.; Pufe, T.; Marx, G.; Fries, M. Argon reduces neurohistopathological damage and preserves functional recovery after cardiac arrest in rats. Br. J. Anaesth. 2013, 110, i106–i112. [Google Scholar] [CrossRef]
  29. Brucken, A.; Kurnaz, P.; Bleilevens, C.; Derwall, M.; Weis, J.; Nolte, K.; Rossaint, R.; Fries, M. Dose dependent neuroprotection of the noble gas argon after cardiac arrest in rats is not mediated by K(ATP)-channel opening. Resuscitation 2014, 85, 826–832. [Google Scholar] [CrossRef] [PubMed]
  30. Brucken, A.; Kurnaz, P.; Bleilevens, C.; Derwall, M.; Weis, J.; Nolte, K.; Rossaint, R.; Fries, M. Delayed argon administration provides robust protection against cardiac arrest-induced neurological damage. Neurocrit. Care 2014. [Google Scholar] [CrossRef]
  31. Fahlenkamp, A.V.; Coburn, M.; de Prada, A.; Gereitzig, N.; Beyer, C.; Haase, H.; Rossaint, R.; Gempt, J.; Ryang, Y.M. Expression analysis following argon treatment in an in vivo model of transient middle cerebral artery occlusion in rats. Med. Gas Res. 2014, 4, 11. [Google Scholar] [CrossRef] [PubMed]
  32. Irani, Y.; Pype, J.L.; Martin, A.R.; Chong, C.F.; Daniel, L.; Gaudart, J.; Ibrahim, Z.; Magalon, G.; Lemaire, M.; Hardwigsen, J. Noble gas (argon and xenon)-saturated cold storage solutions reduce ischemia-reperfusion injury in a rat model of renal transplantation. Nephron Extra 2011, 1, 272–282. [Google Scholar] [CrossRef] [PubMed]
  33. Pagel, P.S.; Krolikowski, J.G.; Shim, Y.H.; Venkatapuram, S.; Kersten, J.R.; Weihrauch, D.; Warltier, D.C.; Pratt, P.F., Jr. Noble gases without anesthetic properties protect myocardium against infarction by activating prosurvival signaling kinases and inhibiting mitochondrial permeability transition in vivo. Anesth. Analg. 2007, 105, 562–569. [Google Scholar] [CrossRef] [PubMed]
  34. Ryang, Y.M.; Fahlenkamp, A.V.; Rossaint, R.; Wesp, D.; Loetscher, P.D.; Beyer, C.; Coburn, M. Neuroprotective effects of argon in an in vivo model of transient middle cerebral artery occlusion in rats. Crit. Care Med. 2011, 39, 1448–1453. [Google Scholar] [CrossRef] [PubMed]
  35. Zhuang, L.; Yang, T.; Zhao, H.; Fidalgo, A.R.; Vizcaychipi, M.P.; Sanders, R.D.; Yu, B.; Takata, M.; Johnson, M.R.; Ma, D. The protective profile of argon, helium, and xenon in a model of neonatal asphyxia in rats. Crit. Care Med. 2012, 40, 1724–1730. [Google Scholar] [CrossRef] [PubMed]
  36. David, H.N.; Haelewyn, B.; Risso, J.J.; Abraini, J.H. Modulation by the noble gas argon of the catalytic and thrombolytic efficiency of tissue plasminogen activator. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2013, 386, 91–95. [Google Scholar] [CrossRef]
  37. Jawad, N.; Rizvi, M.; Gu, J.; Adeyi, O.; Tao, G.; Maze, M.; Ma, D. Neuroprotection (and lack of neuroprotection) afforded by a series of noble gases in an in vitro model of neuronal injury. Neurosci. Lett. 2009, 460, 232–236. [Google Scholar] [CrossRef] [PubMed]
  38. Fahlenkamp, A.V.; Rossaint, R.; Haase, H.; Al Kassam, H.; Ryang, Y.M.; Beyer, C.; Coburn, M. The noble gas argon modifies extracellular signal-regulated kinase 1/2 signaling in neurons and glial cells. Eur. J. Pharmacol. 2012, 674, 104–111. [Google Scholar] [CrossRef] [PubMed]
  39. Galchuk, S.V.; Turovetskii, V.B.; Andreev, A.I.; Buravkova, L.B. Effect of argon and nitrogen on the peritoneal macrophages in mice and their resistance to the UV damaging effect in vitro. Aerosp. Environ. Med. 2001, 35, 39–43. [Google Scholar]
  40. Harris, K.; Armstrong, S.P.; Campos-Pires, R.; Kiru, L.; Franks, N.P.; Dickinson, R. Neuroprotection against traumatic brain injury by xenon, but not argon, is mediated by inhibition at the N-methyl-d-aspartate receptor glycine site. Anesthesiology 2013, 119, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
  41. Loetscher, P.D.; Rossaint, J.; Rossaint, R.; Weis, J.; Fries, M.; Fahlenkamp, A.; Ryang, Y.M.; Grottke, O.; Coburn, M. Argon: Neuroprotection in in vitro models of cerebral ischemia and traumatic brain injury. Crit. Care 2009, 13, R206. [Google Scholar] [CrossRef] [Green Version]
  42. Maio, D.A.; Neville, J.R. Effect of chemically inert gases on oxygen consumption in living tissues. Aerosp. Med. 1967, 38, 1049–1056. [Google Scholar] [PubMed]
  43. Spaggiari, S.; Kepp, O.; Rello-Varona, S.; Chaba, K.; Adjemian, S.; Pype, J.; Galluzzi, L.; Lemaire, M.; Kroemer, G. Antiapoptotic activity of argon and xenon. Cell Cycle 2013, 12, 2636–2642. [Google Scholar] [CrossRef] [PubMed]
  44. Yarin, Y.M.; Amarjargal, N.; Fuchs, J.; Haupt, H.; Mazurek, B.; Morozova, S.V.; Gross, J. Argon protects hypoxia-, cisplatin- and gentamycin-exposed hair cells in the newborn rat’s organ of Corti. Hear. Res. 2005, 201, 1–9. [Google Scholar] [CrossRef] [PubMed]
  45. David, H.N.; Haelewyn, B.; Rouillon, C.; Lecoq, M.; Chazalviel, L.; Apiou, G.; Risso, J.J.; Lemaire, M.; Abraini, J.H. Neuroprotective effects of xenon: A therapeutic window of opportunity in rats subjected to transient cerebral ischemia. FASEB J. 2008, 22, 1275–1286. [Google Scholar] [CrossRef] [PubMed]
  46. Sanders, R.D.; Godlee, A.; Fujimori, T.; Goulding, J.; Xin, G.; Salek-Ardakani, S.; Snelgrove, R.J.; Ma, D.; Maze, M.; Hussell, T. Benzodiazepine augmented gamma-amino-butyric acid signaling increases mortality from pneumonia in mice. Crit. Care Med. 2013, 41, 1627–1636. [Google Scholar] [CrossRef] [PubMed]
  47. Dickinson, R.; Peterson, B.K.; Banks, P.; Simillis, C.; Martin, J.C.; Valenzuela, C.A.; Maze, M.; Franks, N.P. Competitive inhibition at the glycine site of the N-methyl-D-aspartate receptor by the anesthetics xenon and isoflurane: Evidence from molecular modeling and electrophysiology. Anesthesiology 2007, 107, 756–767. [Google Scholar] [CrossRef] [PubMed]
  48. Colloc’h, N.; Marassio, G.; Prange, T. Protein-noble gas interactions investigated by crystallography on three enzymes—implication on anesthesia and neuroprotection mechanisms. In Current Trends in X-ray Crystallography; Chandrasekaran, Annamalai, Ed.; InTech: Rijeka, Croatia, 2011. [Google Scholar]
  49. Quillin, M.L.; Breyer, W.A.; Griswold, I.J.; Matthews, B.W. Size versus polarizability in protein-ligand interactions: Binding of noble gases within engineered cavities in phage T4 lysozyme. J. Mol. Biol. 2000, 302, 955–977. [Google Scholar] [CrossRef] [PubMed]
  50. Soviet’s first Nobelist. In New Scientist; New Science Publications: London, UK, 19 September 1974; Volume 63, Number 915.

Share and Cite

MDPI and ACS Style

Höllig, A.; Schug, A.; Fahlenkamp, A.V.; Rossaint, R.; Coburn, M.; Argon Organo-Protective Network. Argon: Systematic Review on Neuro- and Organoprotective Properties of an “Inert” Gas. Int. J. Mol. Sci. 2014, 15, 18175-18196. https://doi.org/10.3390/ijms151018175

AMA Style

Höllig A, Schug A, Fahlenkamp AV, Rossaint R, Coburn M, Argon Organo-Protective Network. Argon: Systematic Review on Neuro- and Organoprotective Properties of an “Inert” Gas. International Journal of Molecular Sciences. 2014; 15(10):18175-18196. https://doi.org/10.3390/ijms151018175

Chicago/Turabian Style

Höllig, Anke, Anita Schug, Astrid V. Fahlenkamp, Rolf Rossaint, Mark Coburn, and Argon Organo-Protective Network (AON). 2014. "Argon: Systematic Review on Neuro- and Organoprotective Properties of an “Inert” Gas" International Journal of Molecular Sciences 15, no. 10: 18175-18196. https://doi.org/10.3390/ijms151018175

Article Metrics

Back to TopTop