Abstract
Central nervous system injuries are a leading cause of death and disability worldwide. Although the exact pathophysiological mechanisms of various brain injuries vary, central nervous system injuries often result in an inflammatory response, and subsequently lead to brain damage. This suggests that neuroprotection may be necessany in the treatment of multiple disease models. The use of medical gases as neuroprotective agents has gained great attention in the medical field. Medical gases include common gases, such as oxygen, hydrogen and carbon dioxide; hydrogen sulphide and nitric oxide that have been considered toxic; volatile anesthetic gases, such as isoflurane and sevoflurane; and inert gases like helium, argon, and xenon. The neuroprotection from these medical 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. Nevertheless, the transition into the clinical practice is still lagging. This delay could be attributed to the contradictory paradigms and the conflicting results that have been obtained from experimental models, as well as the presence of inconsistent reports regarding their safety. In this review, we summarize the potential mechanisms underlying the neuroprotective effects of medical gases and discuss possible candidates that could improve the outcomes of brain injury.
Keywords: hydrogen, hydrogen sulphide, hyperbaric oxygen, inert gases, nitric oxide, isoflurane, sevoflurane, traumatic brain injury, ischemia/reperfusion, subarachnoid hemorrhage
INTRODUCTION
Central nervous system injuries are a leading cause of death and disability worldwide.1 In China, brain injury is one of the major causes of clinical mortality and long-term disability.2 With China’s economic development, an increase in traffic accidents and brain trauma has been observed over the past few years.2,3,4 Stroke is another health burden in China as it accounts for 80% of deaths and 70% of disability-adjusted life-years lost.5 Additionally, the growth in the aging population of China has led to an increase in the occurrence of neurodegenerative diseases.6,7 Interestingly, brain injuries often share similar underlying pathophysiological mechanisms.1 Therefore, reducing neural damage through the use of neuroprotective agents will improve the patient prognosis.8,9
Medical gases are increasingly used clinically because of their special physicochemical properties and convenience in use. Medical gases exert unique neuroprotective effects against brain injury. Administration of hyperbaric oxygen (HBO) has been successfully used for the treatment of some neurological disorders.10,11 Furthermore, hydrogen (H2) is emerging as an antioxidant agent with neuroprotective properties.12,13,14 Hypercapnia, induced by a high concentration of carbon dioxide, has been demonstrated to be beneficial in the treatment of ischemic brain injuries.15,16,17 Hydrogen sulphide (H2S) and nitric oxide (NO), which were once considered toxic, can act as signaling molecules with a promising role in brain protection.18,19 In addition, clinical studies have demonstrated that inhaled anesthetics, as well as rare gases, provide a certain degree of neuroprotection.20,21 In this review, we searched for neuroprotective studies on common medical gases in the past 5 years, and briefly summarized their application and mechanisms in various neurological diseases, with gas species as a classification criterion (Table 1). It can be seen that the various medical gases reviewed play a protective role in nerve injuries, and the molecular mechanism still has a broad space for exploration. Further, we will discuss the advantages of their use and challenge their wide clinical applications.
Table 1.
Medical gas | Condition | Mechanism of action | Reference |
---|---|---|---|
Hyperbaric oxygen | Traumatic brain injury | IL-10, caspases-3, Bcl-2 | Chen et al.22 |
TNF-a, TGIF, TGF-β1 | Wee et al.23 | ||
NAA/Cr ratio, Cho/Cr ratio | Zhang et al.24 | ||
Middle cerebral artery occlusion | Cleaved caspase-3 | Lu et al.25 | |
Cerebral malaria | Indoleamine 2,3-dioxygenase 1, AhR | Bastos et al.26 | |
Posttraumatic stress disorder | Glucocorticoid receptor | Lin et al.11 | |
Hydrogen | Ischemia/reperfusion | Reactive oxygen species | Ohsawa et al.12 |
Hypoxia/ischemia | caspase-3, caspase-12 | Cai et al.27 | |
Alzheimer’s disease | JNK, NF-κB | Wang et al.28 | |
Traumatic brain injury | Reactive oxygen species | Ji et al.29 | |
IL-1β, IL-10, HMGB1 | Tian et al.30 | ||
Subarachnoid hemorrhage | NF-κB, NLRP3 | Shao et al.31 | |
Cognitive impairment | Estrogen, ERb, BDNF | Hou et al.32 | |
Ischemia/reperfusion | 8-OHdG, reactive oxygen species | Nagatani et al.33 | |
Tregs, miR-21, miR-210 | Li et al.34 | ||
Intracerebral hemorrhage | Reactive oxygen species | Manaenko et al.35 | |
Spontaneously hypertensive stroke-prone | Reactive oxygen species, MMP-9 | Takeuchi et al.36 | |
Brain injury | PI3K/Akt/GSK3β | Chen et al.37 | |
Parkinson’s disease | ghrelin | Yoshii et al.38 | |
Major depressive disorder | IL-1β, reactive oxygen species | Zhang et al.39 | |
Carbon dioxide | Hypoxia/ischemia | AQP4 | Yang et al.15 |
Ischemia/reperfusion | cyt-c, cleaved caspase-3 | Tao et al.17 | |
Bcl-2, Bax | Tao et al.17 | ||
AQP-4, caspase-3 | Zhou et al.40 | ||
Hydrogen sulphide | Middle cerebral artery occlusion | AQP4, PKC | Wei et al.18 |
Subarachnoid hemorrhage | AQP4, MMP-9 | Cao et al.41 | |
IL-1β, CBS, 3MST | Cui et al.42 | ||
Akt/ERK, BDNF-CREB | Li et al.43 | ||
Intracerebral hemorrhage | P2X7R/NLRP3 | Zhao et al.44 | |
Cognitive impairment | Cleaved caspase-3 | Hu et al.45 | |
GluN2B, NMDAR | Zhan et al.46 | ||
Traumatic brain injury | Beclin-1-Vps34 | Zhang et al.47 | |
Parkinson’s disease | ROCK2, miR-135a-5p | Liu et al.48 | |
Nitric oxide | Subarachnoid hemorrhage | Pial arteriole | Terpolilli et al.19 |
Traumatic brain injury | Resistance vessel, CBF | Terpolilli et al.49 | |
Cerebral ischemia | Pial venule, arteriole | Terpolilli et al.50 | |
Ischemia/reperfusion | CBF | Li et al.51 | |
Isoflurane | Ischemia/reperfusion | JNK | Wang et al.20 |
BMP4/Smad1/5/8 | Yuan et al.52 | ||
Cerebral ischemia | Notch | Yin et al.53 | |
Sevoflurane | Hypoxia/reoxygenation | VEGF | Restin et al.54 |
Ischemia/reperfusion | Lysosomal cathepsin B | Zhu et al.55 | |
Cerebral ischemia | TLR-4/NF-κB | Hwang et al.56 | |
Microglia, macrophage | Dang et al.57 | ||
Hypoxia/ischemia | PI3K/Akt-mPTP | Lai et al.58 | |
Hemorrhage shock and resuscitation | GRP78, CHOP | Hu et al.59 | |
Helium | Hypoxia/ischemia | Ang-1, Tie-2, Flt-1 | Li et al.60 |
Argon | Hypoxia/ischemia | PI3K/Akt/HO-1 | Zhao et al.61 |
Subarachnoid hemorrhage | HO-1 | Höllig et al.62 | |
Xenon | Parkinson’s disease | Astrocytes | Lavaur et al.63 |
Ischemic/anoxic | Cholinergic traits | Lavaur et al.64 | |
Traumatic brain injury | NMDAR, TREK-1 | Harris et al.65 | |
Subarachnoid hemorrhage | Microglial | Veldeman et al.66 |
Note: IL-10: Interleukin-10; TNF-a: tumor necrosis factor-a; TGIF: transforming growth interacting factor; TGF-β1: transforming growth factor-beta1; NAA: nonessential amino acid; Cr: creatinine; Cho: cholesterol; AhR: aryl hydrocarbon receptor; JNK: c-Jun N-terminal kinase; NF-κB: nuclear factor-kappa B; IL-1β: interleukin-1β; HMGB1: high mobility group box 1 protein; NLRP3: receptor family pyrin domain-containing 3; ERb: estrogen receptor b; BDNF: brain-derived neurotrophic factor; 8-OHdG: 8-hydroxy-2 deoxyguanosine; miR: microRNA; Tregs: regulatory T cells; MMP-9: matrix metalloproteinase-9; PI3K: phosphatidylinositol 3-hydroxy kinase; Akt: protein kinase B; GSK3β: glycogen synthase kinase 3β; AQP4: aquaporin 4; cyt-c: cytochrome c; PKC: protein kinase C; CBS: cystathionine beta-synthase; 3MST: 3-mercaptopyruvate sulfur transferase; ERK: extracellular regulated protein kinases; CREB: cyclic adenosine monophosphate-response element binding protein; P2X7R: P2X7 receptor; GluN2B: phospho-NMDA receptor 2B; NMDAR: N-methyl-D-aspartate receptor; Vps34: phosphatidylinositol 3-kinase; ROCK2: Rho-associated protein kinase 2; CBF: cerebral blood flow; BMP4: bone morphogenetic protein 4; Smad: mothers against decapentaplegic homolog; VEGF: vascular endothelial growth factor; TLR-4: Toll-like receptor 4; mPTP: mitochondrial permeability transition pore; GRP78: glucose-regulated protein-78; CHOP: C/EBP-homologous protein; Ang-1: angiopoietin-1; Tie-2: tyrosine kinase with immunoglobulin and epidermalgrowth factor homology domains-2; Flt-1: vascular endothelial growth factor receptor 1; HO-1: heme oxygenase 1; TREK-1: TWIK-related K+ channel 1.
HYPERBARIC OXYGEN
HBO treatment refers to the inhalation of pure oxygen, or a high concentration of oxygen, in a high pressure environment to treat hypoxic conditions.67 HBO is the main treatment for carbon monoxide poisoning and decompression sickness.68 Recently, HBO has become an issue of concern for its beneficial effects in the treatment of brain injuries.
Chen et al.22 and Wee et al.23 previously confirmed that inflammation plays an important role in the pathophysiology of traumatic brain injuries (TBI) in animal models. Following TBI, a lack of interleukin (IL)-10 counters the protective effect of HBO thereby increasing the degree of brain injury.22 Therefore, this anti-inflammatory cytokine (IL-10) plays a crucial role in mediating the neuroprotective effect of HBO. The immediate inhalation of HBO (2.0 atmosphere absolute (ATA), 1 ATA = 1.013 kPa, 100% O2) after brain injury decreases apoptosis25 and reduces the expression of inflammatory cytokines.23,24,26 Moreover, HBO has been found to protect the integrity of the blood-brain barrier (BBB) and improve the patient prognosis.24,69 Lin et al.11 also demonstrated the positive impact of HBO administration on the behavioral and neurochemical outcomes of posttraumatic stress disorder. In mice, the continuous inhalation of HBO immediately after TBI reduces neural loss and increases the activity of astrocytes.10 Moreover, continuous HBO inhalation has been shown to exert a more significant neuroprotective effect than non-continuous inhalation.10
Clinical trials have shown that inhalation of HBO (2.0 ATA, 100% O2) promotes the regeneration of cerebral blood vessels and the reconstruction of nerve fibers after TBI.9 In healthy volunteers, the inhalation of HBO significantly enhances the cognitive functions and ability to perform cognitive tasks compared to the performance following inhalation of normobaric air.70 In addition, long-term HBO therapy could improve post-concussion syndrome and post-traumatic stress disorder after moderate brain injury and may significantly reduce post-traumatic anxiety and suicide. Nevertheless, at present, the impact of HBO has been derived from small and uncontrolled studies or single case reports. Therefore, randomized double-blinded clinical trials are required to enable the wide clinical application of HBO.
HYDROGEN
H2 gas is chemically stable at room temperature. It has a small molecular weight and strong permeability, which enables its diffusion into the cells. In 1975, hyperbaric H2 therapy was proposed as a possible anticancer agent.71 However, the selective anti-oxidative effects of H2 were discovered more recently, in 2007.12 Ohsawa et al.12 reported that H2 significantly reduces the area of cerebral infarction by neutralizing toxic free radicals. Subsequent research, performed in experimental animals, found that H2 has anti-inflammatory,69,72 anti-apoptotic,27,73 and antioxidant28 properties. Additionally, H2 has been shown to have neuroprotective effects in cerebrovascular diseases and neurodegenerative diseases.32,33,35 Takeuchi et al.36 demonstrated that drinking hydrogen water could reduce the production of reactive oxygen species and inhibit the activation of matrix metalloproteinase-9 in the hippocampus, thereby reducing BBB damage and improving brain function. In a hypoxic-ischemic encephalopathy piglet model, treatment with H2 reduced oxidative stress and improved neural recovery.74
Cerebral microvascular endothelial cells play an important role in regulating and maintaining the stability and balance of the brain neurovascular microenvironment.37 Consuming hydrogen water has been shown to prevent the apoptosis of cerebral microvascular endothelial cells through the down-regulation of the phosphatidylinositol 3-hydroxy kinase/protein kinase B/glycogen synthase kinase 3β (PI3K/Akt/Gsk3β) pathway, leading to a decrease in the extent of secondary brain injury.37 Systemic and central nervous system inflammation induces microglial activation, causing neuronal injury. H2 inhibits microglial activation, thus protecting against brain trauma.30 In a rat model of subarachnoid hemorrhage, injection of H2 saline inhibits the nuclear factor-kappa B pathway and the nucleotide binding and oligomerization domain-like receptor family pyrin domain–containing three inflammasomes, which subsequently reduces the systemic inflammatory response and promotes the neurological function, as well as behavioural recovery.31 Yoshii et al.38 reported that the observed neuroprotective effect of drinking hydrogen water could be associated with the secretion of ghrelin, a gastric hormone, in the stomach. However, the mechanism of H2 appears to differ between the various experimental models. For instance, H2 inhibits the immuno-inflammatory response by up-regulating the expression of regulatory T cells after cerebral ischemia/reperfusion injury.34 Activation of inflammatory mediators plays an important role in major depressive disorder.39 Hydrogen water inhibits the production of IL-1β and reactive oxygen species, reduces the inflammatory reaction, and subsequently decreases the depression behavioural scores.39 In a model of cerebral infarction, glutamate producesdose-dependent neurocytotoxicity and increases the intracellular calcium level.75 The neuroprotective function of hydrogen water, in this case, was found to be mediated through a reduction in glutamate-induced neural cell death and inhibition of calcium ion influx.75 On the other hand, treatment with H2 failed to relieve brain edema or exert a neuroprotective function, although it did decrease the expression of 8-hydroxy-2′-deoxyguanosine, in a rat model of intracerebral hemorrhage. This result may be attributed to the low H2 concentration that was used in this study, or to the existence of a more complicated mechanism involving reactive oxygen species in intracerebral hemorrhage.76 Therefore, the protective effects of H2 may differ according to the dosage and/or animal model that are used. Nevertheless, H2 is safe to the human body at a high concentration and it plays an anti-inflammatory, as well as an antioxidant, role in combating cerebral ischemia/reperfusion injury.13 Therefore, the protective effects of H2 in TBI are dose- and time-dependent. Indeed, we observed that the inhalation of a high H2 concentration exerts neuroprotective effects against TBI in diabetic mice (unpublished observations). Taken together, these findings indicate that the neuroprotective effect of H2 must be replicated in clinical studies to enable its future application.
CARBON DIOXIDE
Carbon dioxide is a liposoluble gas that can cross the cell membrane and the BBB.77 Therapeutic hypercapnia, through the inhalation of carbon dioxide, has been shown to exert beneficial biological functions.78,79,80 The use of hypercapnia in organ protection has been the focus of recent research especially its effect on the brain function.15,16 Hypercapnia significantly reduces the infarct size and improves the neuropathologic score (neurosensitivity, reflex, and exercise behaviour) in a model of cerebral ischemic injury.16 In addition, hypercapnia enhances the spatial memory and improves the sensorimotor impairment by regulating apoptosis, through an up-regulation of Bcl-2 and a down-regulation of Bax.17 In a control case study, controlled transient hypercapnia increased the cerebral blood flow and cerebral oxygen saturation and reduced the possibility of secondary cerebral infarction in patients without adverse reactions.81 In a rat model of global cerebral ischemia/reperfusion, Zhou et al.40 reported that moderate hypercapnia (partial pressure of carbon dioxide (PaCO2) 80–100 mmHg) had significantly improved neuroprotective effects compared to those of mild hypercapnia (PaCO2 60–80 mmHg); whereas severe hypercapnia (PaCO2 100–120 mmHg) increased the severity of brain injury. In agreement with these findings, Yang et al.15 reported that mild-to-moderate hypercapnia (PaCO2 60–80 mmHg and PaO2 > 50 mmHg) was the optimal concentration range for neuroprotective effects. At this level, hypercapnia significantly reduces the BBB permeability as well as the brain water content, the expression of Aquaporin-4 (AQP4) and neural apoptosis.15 Conversely, hypercapnia in combination with lower oxygen pressure (PaO2 < 50 mmHg) may aggravate BBB destruction and edema due to cerebral ischemia/hypoxia. Therefore, the protective effect of carbon dioxide in the brain is determined by the degree of hypercapnia as well as the partial pressure of oxygen.15 In our experience, we have confirmed that hypercapnia (3 hours with PaCO2 levels of 80–100 mmHg) reduces brain edema, improves the BBB function, reduces the lesion volume, and improves the neurological outcome in a rat model of fluid percussion injury (unpublished observations). Taken together, these results suggest that there is a potential neuroprotective role for “therapeutic hypercapnia” after brain injury. However, confirming these results through the use of experimental animal models will be instrumental in elucidating the neuroprotective mechanisms of hypercapnia and acidosis before conducting clinical trials.
HYDROGEN SULPHIDE
Hydrogen sulphide (H2S), an endogenous gaseous signalling conduction factor, was considered to be a toxic substance for quite a long period of time. However, recent studies have confirmed that low doses of H2S play an important role in the functioning of normal physiological processes.82,83,84 In fact, H2S was found to be widely involved in physiological and pathophysiological processes in the brain as well as the peripheral tissues.82 Following brain injury accompanied with brain edema, the expression of AQP4 increases remarkably.18,41 Accumulating evidence has shown that the neuroprotective impact of H2S is mediated by its ability to increase the expression of protein kinase C and decrease the expression of matrix metalloprotein-9 through the inhibition of AQP4 expression, which in turn leads to the suppression of glial cell activation and the release of pro-inflammatory factors.18,41,44 In rats, H2S relieves cerebral vasospasm, protects neurons, preserves endothelial function, reduces cerebral edema, and inhibits apoptosis as well as the inflammatory response.42,44 Additionally, Zhang et al.47 demonstrated that the administration of H2S attenuates apoptosis, inhibits the activation of autophagy and improves motor function following TBI. H2S could also improve the learning and cognitive ability as well as preserving the memory functions after brain injury.46 Taken together, these results indicate that H2S can improve the sequelae of brain injury, including long-term disability.43,85 Li et al.43 suggested that the neuroprotective impact of H2S could be mediated through the induction of the Akt-ERK pathway. Rho-associated protein kinase 2 is a key factor that promotes neurodegeneration in Parkinson’s disease.48 H2S could reduce the expression of Rho-associated protein kinase 2 through microRNA-mediated protection of nerve cells.48 Future research should focus on uncovering the exact role of H2S in the central nervous system with the aims of dissecting the signaling pathways involved.
NITRIC OXIDE
Like H2S, NO has previously been considered to be a toxic chemical substance.86 In the 1980s, the vasodilating factor secreted by vascular endothelial cells was identified as NO.87 Recent reports have suggested that NO inhalation successfully reduces the size of the necrotic area and brain edema as well as reduces BBB permeability following subarachnoid hemorrhage or TBI.19,49 Thus, NO may improve neurological function and relieve secondary brain injury.49 Additionally, NO inhalation may alleviate the spasm of pia mater arterioles,50 improve the neurological score, and reduce the mortality rate in mice.19 The concentration of NO and inhalation time were positively correlated with its neuroprotective effects.51 The neuroprotective function of NO is mediated through the improvement of the cerebral blood flow without causing hypotension or other significant side effects.51 At present, evidence for the neuroprotective impact of NO is based on the results of animal experiments. Clinical studies are required to confirm the role of NO in TBI. Additionally, research studies examining the mechanism of action of NO as well as the toxicity and side effects of long-term use will influence the future therapeutic applications of NO.
VOLATILE ANESTHETIC GASES
Isoflurane
Isoflurane is a general anesthetic mainly used to start or maintain anesthesia. Following a cerebral ischemic event, microglial activation may induce neural apoptosis in the brain.53,88 Isoflurane has been found to inhibit the activation of microglia through the Notch pathway, therefore, producing a decrease in apoptosis.53 Additionally, Wang et al.20 and Yuan et al.52 demonstrated that isoflurane could alleviate the incidence of brain edema and reduce the area of reperfusion injury by down-regulating the expression of AQP4. Moreover, isoflurane inhalation up-regulates transforming growth factor-beta1 expression and down-regulates phospho-c-Jun N-terminal kinase expression, leading to an improvement in the ischemia/reperfusion injury outcome.20 However, in cases of severe brain injury, long-interval isoflurane inhalation may reverse its previous protective effect and aggravate brain injury.89 Therefore, future studies should focus on optimizing the ideal dose and time-frame for isoflurane inhalation to lay a solid foundation for the widespread application of isoflurane in the treatment of TBI.
Sevoflurane
To date, sevoflurane has been considered to be an ideal inhalation anaesthetic, owing to its rapid induction and revival.55 In vitro experiments have demonstrated that sevoflurane can down-regulate the expression of vascular endothelial growth factor, maintain the function of the endothelial barrier, and play a key role in injury regulation.54 Cerebral ischemia often leads to the astrocyte activation and the formation of a glial scar.55 In addition, sevoflurane inhalation alleviates reactive astrocytic gelatinization, reduces the effect of glial scar formation, and inhibits the activation and release of lysosomal cathepsin B, which improves the outcome of cerebral ischemia.55 Sevoflurane may also attenuate the inflammatory response.56,88 However, this anesthetic neither relieves inflammation in the brain nor significantly inhibits the activation of microglia or astrocytes.88 Nevertheless, Dang et al.57 demonstrated that the neuroprotective impact of sevoflurane could be attributed to the activation of microglia/macrophage migration and, thus, the promotion of brain repair. Additionally, sevoflurane post-conditioning improves the cognitive performance in rats as well as promoting neural survival and decreasing apoptosis and cellular atrophy through the regulation of the PI3K/Akt pathway.58,90 Conversely, several studies have reported that sevoflurane exhibit neurotoxic effects.21,91,92,93 Inhalation of 7% sevoflurane in aged rats inhibits the expression of brain-derived neurotrophic factor, aggravates postoperative cognitive dysfunction and impairs the brain function.21 Moreover, sevoflurane-induced nerve injury is correlated with the used concentration.91 Therefore, future research should explore the neuroprotective mechanisms of sevoflurane and optimize the concentration that is selected for research. Nevertheless, the neuroprotective effect of sevoflurane provides unique opportunities for its application by clinical anesthesiologists in cases of TBI.
INERT GASES
Helium
Helium has a lower solubility in blood than nitrogen and is often mixed with oxygen to provide a breathing gas for divers.94 Angiogenesis is a natural defense mechanism that provides oxygen and nutrient supply to the injured brain.60 Li et al.60 demonstrated that helium exerts its neuroprotective effects by improving the neurovascular niche, as well as increasing the expression of anti-inflammatory cytokines and BNDF. However, Aehling et al.95 did not observe any beneficial effect of helium preconditioning or post-conditioning on neurological function. However, it did reduce the level of apoptosis in a rat resuscitation model.95 Future studies will improve our understanding of the function of helium in neuroprotection.
Argon
The neuroprotective effects of argon have been validated in various brain injury models.96 Argon improves the general condition of rats and reduces mortality by inducing the expression of Heme oxygenase 1.61,62 Compared to the impact of argon alone, the combination of argon and hypothermia therapy significantly increases the expression of Heme oxygenase 1, reduces the infarct size, and provides effective protection against short-term and long-term brain injury.61 Although there are still many uncertainties regarding the effect and mechanism of argon therapy, its neuroprotective role in TBI is worth further exploration.
Xenon
Xenon is an ideal anesthetic. However, the widespread clinical application of xenon is hampered by the difficulty of its separation and its scarce availability. Nevertheless, the potential neuroprotective effect of xenon has been the focus of scientific research.97 Xenon alleviates oxidative stress, reduces N-methyl-D-aspartate receptor mediated neurodegeneration and exerts both neurotrophic as well as neuroprotective effects in cholinergic neurons.64,65 Additionally, xenon produces neuroprotection by inhibiting the activation of microglia and reducing the level of hippocampal neural damage.66 In Parkinson’s disease, xenon may protect and nourish dopamine neurons, thus inhibiting the potential damage to dopaminergic neurons and astrocytes.63 However, the feasibility of the widespread clinical application of xenon remains to be explored in future experiments.
PERSPECTIVES
The debate regarding the neuroprotective role of medical gases is still ongoing. To date, there are significant differences in the observed value of medical gases across different studies. However, the evidence from published studies suggests that each of the medical gases discussed in this review exert different degrees of brain protection in specific nerve injury models. It is worth mentioning that, although these gases have different uses in various types of brain injuries, the mechanisms by which these gases reduce neuronal injury at the intracellular and intercellular level are similar. The major challenge that faces this field of research is the translation from preclinical to clinical training, i.e., the feasibility of the clinical administration of medical gases and their inclusion in individualized treatment plans. HBO should be considered to be a supportive therapeutic modality in TBI. The use of carbon dioxide in neuroprotective therapy is still at the experimental animal stage and, thus, there is an urgent need for more extensive research to determine its therapeutic value. The lack of precise methodology for the accurate estimation of H2S and NO endogenous concentrations hinders their clinical use. Volatile anesthetic gases may not be effective as single therapeutic agents. However, their use might be beneficial during neuroprotective surgeries. The clinical administration of inert gases is hampered by high costs and difficulty of their extraction. H2 is a popular gas with definite roles in disease prevention and treatment and has a high safety margin. These characteristics imply that H2 may revolutionize the medical field in the future. Future research should aim to clarify the specific mechanisms that underlie the action of H2. In conclusion, in this review we present recent insights into the therapeutic uses and possible applications of medical gases. Future research will focus on the precise function and molecular mechanism of each medical gas, which will lay a solid foundation for their widespread application in clinical practice.
Footnotes
Funding: This work was supported by the National Natural Science Foundation of China, No. 81400989 (to WCY).
Conflicts of interest
The authors declare no conflicts of interest.
Financial support
This work was supported by the National Natural Science Foundation of China, No. 81400989 (to WCY).
Copyright license agreement
The Copyright License Agreement has been signed by all authors before publication.
Plagiarism check
Checked twice by iThenticate.
Peer review
Externally peer reviewed.
REFERENCES
- 1.Anthony DC, Couch Y. The systemic response to CNS injury. Exp Neurol. 2014;258:105–111. doi: 10.1016/j.expneurol.2014.03.013. [DOI] [PubMed] [Google Scholar]
- 2.Cheng P, Yin P, Ning P, et al. Trends in traumatic brain injury mortality in China, 2006-2013: A population-based longitudinal study. PLoS Med. 2017;14:e1002332. doi: 10.1371/journal.pmed.1002332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Li Y, Chen F, Zhang J, et al. Epidemiological characteristics of Chinese paediatric traumatic brain injury inpatients. Brain Inj. 2017;31:1094–1101. doi: 10.1080/02699052.2017.1298004. [DOI] [PubMed] [Google Scholar]
- 4.Wu X, Hu J, Zhuo L, et al. Epidemiology of traumatic brain injury in eastern China, 2004: a prospective large case study. J Trauma. 2008;64:1313–1319. doi: 10.1097/TA.0b013e318165c803. [DOI] [PubMed] [Google Scholar]
- 5.Wang W, Jiang B, Sun H, et al. Prevalence, incidence, and mortality of stroke in China: results from a nationwide population-based survey of 480 687 adults. Circulation. 2017;135:759–771. doi: 10.1161/CIRCULATIONAHA.116.025250. [DOI] [PubMed] [Google Scholar]
- 6.Chan KY, Wang W, Wu JJ, et al. Epidemiology of Alzheimer’s disease and other forms of dementia in China, 1990-2010: a systematic review and analysis. Lancet. 2013;381:2016–2023. doi: 10.1016/S0140-6736(13)60221-4. [DOI] [PubMed] [Google Scholar]
- 7.Sun Q, Wang T, Jiang TF, et al. Effect of a leucine-rich repeat kinase 2 variant on motor and non-motor symptoms in Chinese Parkinson’s disease patients. Aging Dis. 2016;7:230–236. doi: 10.14336/AD.2015.1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nicolson GL, de Mattos GF, Settineri R, et al. Clinical effects of hydrogen administration: from animal and human diseases to exercise medicine. Int J Clin Med. 2016;7:32–76. [Google Scholar]
- 9.Tal S, Hadanny A, Sasson E, Suzin G, Efrati S. Hyperbaric oxygen therapy can induce angiogenesis and regeneration of nerve fibers in traumatic brain injury patients. Front Hum Neurosci. 2017;11:508. doi: 10.3389/fnhum.2017.00508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Baratz-Goldstein R, Toussia-Cohen S, Elpaz A, Rubovitch V, Pick CG. Immediate and delayed hyperbaric oxygen therapy as a neuroprotective treatment for traumatic brain injury in mice. Mol Cell Neurosci. 2017;83:74–82. doi: 10.1016/j.mcn.2017.06.004. [DOI] [PubMed] [Google Scholar]
- 11.Lin CC, Huang KL, Tung CS, Liu YP. Hyperbaric oxygen therapy restored traumatic stress-induced dysregulation of fear memory and related neurochemical abnormalities. Behav Brain Res. 2019;359:861–870. doi: 10.1016/j.bbr.2018.07.014. [DOI] [PubMed] [Google Scholar]
- 12.Ohsawa I, Ishikawa M, Takahashi K, et al. Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat Med. 2007;13:688–694. doi: 10.1038/nm1577. [DOI] [PubMed] [Google Scholar]
- 13.Cui J, Chen X, Zhai X, et al. Inhalation of water electrolysis-derived hydrogen ameliorates cerebral ischemia-reperfusion injury in rats - A possible new hydrogen resource for clinical use. Neuroscience. 2016;335:232–241. doi: 10.1016/j.neuroscience.2016.08.021. [DOI] [PubMed] [Google Scholar]
- 14.Diao M, Zhang S, Wu L, et al. Hydrogen gas inhalation attenuates seawater instillation-induced acute lung injury via the Nrf2 pathway in rabbits. Inflammation. 2016;39:2029–2039. doi: 10.1007/s10753-016-0440-1. [DOI] [PubMed] [Google Scholar]
- 15.Yang W, Zhang X, Wang N, et al. Effects of acute systemic hypoxia and hypercapnia on brain damage in a rat model of hypoxia-ischemia. PLoS One. 2016;11:e0167359. doi: 10.1371/journal.pone.0167359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tregub P, Kulikov V, Motin Y, Bespalov A, Osipov I. Combined exposure to hypercapnia and hypoxia provides its maximum neuroprotective effect during focal ischemic injury in the brain. J Stroke Cerebrovasc Dis. 2015;24:381–387. doi: 10.1016/j.jstrokecerebrovasdis.2014.09.003. [DOI] [PubMed] [Google Scholar]
- 17.Tao T, Zhao M, Yang W, Bo Y, Li W. Neuroprotective effects of therapeutic hypercapnia on spatial memory and sensorimotor impairment via anti-apoptotic mechanisms after focal cerebral ischemia/reperfusion. Neurosci Lett. 2014;573:1–6. doi: 10.1016/j.neulet.2014.04.051. [DOI] [PubMed] [Google Scholar]
- 18.Wei X, Zhang B, Cheng L, et al. Hydrogen sulfide induces neuroprotection against experimental stroke in rats by down-regulation of AQP4 via activating PKC. Brain Res. 2015;1622:292–299. doi: 10.1016/j.brainres.2015.07.001. [DOI] [PubMed] [Google Scholar]
- 19.Terpolilli NA, Feiler S, Dienel A, et al. Nitric oxide inhalation reduces brain damage, prevents mortality, and improves neurological outcome after subarachnoid hemorrhage by resolving early pial microvasospasms. J Cereb Blood Flow Metab. 2016;36:2096–2107. doi: 10.1177/0271678X15605848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wang S, Yin J, Ge M, et al. Transforming growth-beta 1 contributes to isoflurane postconditioning against cerebral ischemia-reperfusion injury by regulating the c-Jun N-terminal kinase signaling pathway. Biomed Pharmacother. 2016;78:280–290. doi: 10.1016/j.biopha.2016.01.030. [DOI] [PubMed] [Google Scholar]
- 21.Wang JY, Feng Y, Fu YH, Liu GL. Effect of sevoflurane anesthesia on brain is mediated by lncRNA HOTAIR. J Mol Neurosci. 2018;64:346–351. doi: 10.1007/s12031-018-1029-y. [DOI] [PubMed] [Google Scholar]
- 22.Chen X, Duan XS, Xu LJ, et al. Interleukin-10 mediates the neuroprotection of hyperbaric oxygen therapy against traumatic brain injury in mice. Neuroscience. 2014;266:235–243. doi: 10.1016/j.neuroscience.2013.11.036. [DOI] [PubMed] [Google Scholar]
- 23.Wee HY, Lim SW, Chio CC, Niu KC, Wang CC, Kuo JR. Hyperbaric oxygen effects on neuronal apoptosis associations in a traumatic brain injury rat model. J Surg Res. 2015;197:382–389. doi: 10.1016/j.jss.2015.04.052. [DOI] [PubMed] [Google Scholar]
- 24.Zhang Y, Yang Y, Tang H, et al. Hyperbaric oxygen therapy ameliorates local brain metabolism, brain edema and inflammatory response in a blast-induced traumatic brain injury model in rabbits. Neurochem Res. 2014;39:950–960. doi: 10.1007/s11064-014-1292-4. [DOI] [PubMed] [Google Scholar]
- 25.Lu K, Wang H, Ge X, et al. Hyperbaric oxygen protects against cerebral damage in permanent middle cerebral artery occlusion rats and inhibits autophagy activity. Neurocrit Care. 2019;30:98–105. doi: 10.1007/s12028-018-0577-x. [DOI] [PubMed] [Google Scholar]
- 26.Bastos MF, Kayano A, Silva-Filho JL, et al. Inhibition of hypoxia-associated response and kynurenine production in response to hyperbaric oxygen as mechanisms involved in protection against experimental cerebral malaria. FASEB J. 2018;32:4470–4481. doi: 10.1096/fj.201700844R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Cai J, Kang Z, Liu WW, et al. Hydrogen therapy reduces apoptosis in neonatal hypoxia-ischemia rat model. Neurosci Lett. 2008;441:167–172. doi: 10.1016/j.neulet.2008.05.077. [DOI] [PubMed] [Google Scholar]
- 28.Wang C, Li J, Liu Q, et al. Hydrogen-rich saline reduces oxidative stress and inflammation by inhibit of JNK and NF-kappaB activation in a rat model of amyloid-beta-induced Alzheimer’s disease. Neurosci Lett. 2011;491:127–132. doi: 10.1016/j.neulet.2011.01.022. [DOI] [PubMed] [Google Scholar]
- 29.Ji X, Liu W, Xie K, et al. Beneficial effects of hydrogen gas in a rat model of traumatic brain injury via reducing oxidative stress. Brain Res. 2010;1354:196–205. doi: 10.1016/j.brainres.2010.07.038. [DOI] [PubMed] [Google Scholar]
- 30.Tian R, Hou Z, Hao S, et al. Hydrogen-rich water attenuates brain damage and inflammation after traumatic brain injury in rats. Brain Res. 2016;1637:1–13. doi: 10.1016/j.brainres.2016.01.029. [DOI] [PubMed] [Google Scholar]
- 31.Shao A, Wu H, Hong Y, et al. Hydrogen-rich saline attenuated subarachnoid hemorrhage-induced early brain injury in rats by suppressing inflammatory response: possible involvement of NF-κB pathway and NLRP3 inflammasome. Mol Neurobiol. 2016;53:3462–3476. doi: 10.1007/s12035-015-9242-y. [DOI] [PubMed] [Google Scholar]
- 32.Hou C, Peng Y, Qin C, Fan F, Liu J, Long J. Hydrogen-rich water improves cognitive impairment gender-dependently in APP/PS1 mice without affecting Aβ clearance. Free Radic Res. 2018;52:1311–1322. doi: 10.1080/10715762.2018.1460749. [DOI] [PubMed] [Google Scholar]
- 33.Nagatani K, Wada K, Takeuchi S, et al. Effect of hydrogen gas on the survival rate of mice following global cerebral ischemia. Shock. 2012;37:645–652. doi: 10.1097/SHK.0b013e31824ed57c. [DOI] [PubMed] [Google Scholar]
- 34.Li Q, Yu P, Zeng Q, et al. Neuroprotective effect of hydrogen-rich saline in global cerebral ischemia/reperfusion rats: up-regulated Tregs and down-regulated miR-21, miR-210 and NF-κB expression. Neurochem Res. 2016;41:2655–2665. doi: 10.1007/s11064-016-1978-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Manaenko A, Lekic T, Ma Q, Ostrowski RP, Zhang JH, Tang J. Hydrogen inhalation is neuroprotective and improves functional outcomes in mice after intracerebral hemorrhage. Acta Neurochir Suppl. 2011;111:179–183. doi: 10.1007/978-3-7091-0693-8_30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Takeuchi S, Nagatani K, Otani N, et al. Hydrogen improves neurological function through attenuation of blood-brain barrier disruption in spontaneously hypertensive stroke-prone rats. BMC Neurosci. 2015;16:22. doi: 10.1186/s12868-015-0165-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen K, Wang N, Diao Y, et al. Hydrogen-rich saline attenuates brain injury induced by cardiopulmonary bypass and inhibits microvascular endothelial cell apoptosis via the PI3K/Akt/GSK3beta signaling pathway in rats. Cell Physiol Biochem. 2017;43:1634–1647. doi: 10.1159/000484024. [DOI] [PubMed] [Google Scholar]
- 38.Yoshii Y, Inoue T, Uemura Y, et al. Complexity of stomach-brain interaction induced by molecular hydrogen in Parkinson’s disease model mice. Neurochem Res. 2017;42:2658–2665. doi: 10.1007/s11064-017-2281-1. [DOI] [PubMed] [Google Scholar]
- 39.Zhang Y, Su WJ, Chen Y, et al. Effects of hydrogen-rich water on depressive-like behavior in mice. Sci Rep. 2016;6:23742. doi: 10.1038/srep23742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhou Q, Cao B, Niu L, et al. Effects of permissive hypercapnia on transient global cerebral ischemia-reperfusion injury in rats. Anesthesiology. 2010;112:288–297. doi: 10.1097/ALN.0b013e3181ca8257. [DOI] [PubMed] [Google Scholar]
- 41.Cao S, Zhu P, Yu X, et al. Hydrogen sulfide attenuates brain edema in early brain injury after subarachnoid hemorrhage in rats: Possible involvement of MMP-9 induced blood-brain barrier disruption and AQP4 expression. Neurosci Lett. 2016;621:88–97. doi: 10.1016/j.neulet.2016.04.018. [DOI] [PubMed] [Google Scholar]
- 42.Cui Y, Duan X, Li H, et al. Hydrogen sulfide ameliorates early brain injury following subarachnoid hemorrhage in rats. Mol Neurobiol. 2016;53:3646–3657. doi: 10.1007/s12035-015-9304-1. [DOI] [PubMed] [Google Scholar]
- 43.Li T, Liu H, Xue H, et al. Neuroprotective effects of hydrogen sulfide against early brain injury and secondary cognitive deficits following subarachnoid hemorrhage. Brain Pathol. 2017;27:51–63. doi: 10.1111/bpa.12361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhao H, Pan P, Yang Y, et al. Endogenous hydrogen sulphide attenuates NLRP3 inflammasome-mediated neuroinflammation by suppressing the P2X7 receptor after intracerebral haemorrhage in rats. J Neuroinflammation. 2017;14:163. doi: 10.1186/s12974-017-0940-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hu HY, Cui ZH, Li HQ, et al. Fumanjian, a classic Chinese herbal formula, can ameliorate the impairment of spatial learning and memory through apoptotic signaling pathway in the hippocampus of rats with Abeta 1-40 -induced Alzheimer’s disease. Evid Based Complement Alternat Med. 2014;2014:942917. doi: 10.1155/2014/942917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhan JQ, Zheng LL, Chen HB, et al. Hydrogen sulfide reverses aging-associated amygdalar synaptic plasticity and fear memory deficits in rats. Front Neurosci. 2018;12:390. doi: 10.3389/fnins.2018.00390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang M, Shan H, Chang P, et al. Hydrogen sulfide offers neuroprotection on traumatic brain injury in parallel with reduced apoptosis and autophagy in mice. PLoS One. 2014;9:e87241. doi: 10.1371/journal.pone.0087241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Liu Y, Liao S, Quan H, Lin Y, Li J, Yang Q. Involvement of microRNA-135a-5p in the protective effects of hydrogen sulfide against Parkinson’s disease. Cell Physiol Biochem. 2016;40:18–26. doi: 10.1159/000452521. [DOI] [PubMed] [Google Scholar]
- 49.Terpolilli NA, Kim SW, Thal SC, Kuebler WM, Plesnila N. Inhaled nitric oxide reduces secondary brain damage after traumatic brain injury in mice. J Cereb Blood Flow Metab. 2013;33:311–318. doi: 10.1038/jcbfm.2012.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Terpolilli NA, Kim SW, Thal SC, et al. Inhalation of nitric oxide prevents ischemic brain damage in experimental stroke by selective dilatation of collateral arterioles. Circ Res. 2012;110:727–738. doi: 10.1161/CIRCRESAHA.111.253419. [DOI] [PubMed] [Google Scholar]
- 51.Li YS, Shemmer B, Stone E, M AN, Jonas S, Quartermain D. Neuroprotection by inhaled nitric oxide in a murine stroke model is concentration and duration dependent. Brain Res. 2013;1507:134–145. doi: 10.1016/j.brainres.2013.02.031. [DOI] [PubMed] [Google Scholar]
- 52.Yuan M, Ge M, Yin J, et al. Isoflurane post-conditioning down-regulates expression of aquaporin 4 in rats with cerebral ischemia/reperfusion injury and is possibly related to bone morphogenetic protein 4/Smad1/5/8 signaling pathway. Biomed Pharmacother. 2018;97:429–438. doi: 10.1016/j.biopha.2017.10.082. [DOI] [PubMed] [Google Scholar]
- 53.Yin J, Li H, Feng C, Zuo Z. Inhibition of brain ischemia-caused notch activation in microglia may contribute to isoflurane postconditioning-induced neuroprotection in male rats. CNS Neurol Disord Drug Targets. 2014;13:718–732. doi: 10.2174/1871527313666140618110837. [DOI] [PubMed] [Google Scholar]
- 54.Restin T, Kajdi ME, Schlapfer M, et al. Sevoflurane protects rat brain endothelial barrier structure and function after hypoxia-reoxygenation injury. PLoS One. 2017;12:e0184973. doi: 10.1371/journal.pone.0184973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhu YM, Gao X, Ni Y, et al. Sevoflurane postconditioning attenuates reactive astrogliosis and glial scar formation after ischemia-reperfusion brain injury. Neuroscience. 2017;356:125–141. doi: 10.1016/j.neuroscience.2017.05.004. [DOI] [PubMed] [Google Scholar]
- 56.Hwang JW, Jeon YT, Lim YJ, Park HP. Sevoflurane postconditioning-induced anti-inflammation via Inhibition of the Toll-like receptor-4/nuclear factor kappa b pathway contributes to neuroprotection against transient global cerebral ischemia in rats. Int J Mol Sci. 2017;18:E2347. doi: 10.3390/ijms18112347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Dang DD, Saiyin H, Yu Q, Liang WM. Effects of sevoflurane preconditioning on microglia/macrophage dynamics and phagocytosis profile against cerebral ischemia in rats. CNS Neurosci Ther. 2018;24:564–571. doi: 10.1111/cns.12823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lai Z, Zhang L, Su J, Cai D, Xu Q. Sevoflurane postconditioning improves long-term learning and memory of neonatal hypoxia-ischemia brain damage rats via the PI3K/Akt-mPTP pathway. Brain Res. 2016;1630:25–37. doi: 10.1016/j.brainres.2015.10.050. [DOI] [PubMed] [Google Scholar]
- 59.Hu X, Wang J, Zhang L, Zhang Q, Duan X, Zhang Y. Postconditioning with sevoflurane ameliorates spatial learning and memory deficit via attenuating endoplasmic reticulum stress induced neuron apoptosis in a rat model of hemorrhage shock and resuscitation. Brain Res. 2018;1696:49–55. doi: 10.1016/j.brainres.2018.05.047. [DOI] [PubMed] [Google Scholar]
- 60.Li Y, Zhang P, Liu Y, Liu W, Yin N. Helium preconditioning protects the brain against hypoxia/ischemia injury via improving the neurovascular niche in a neonatal rat model. Behav Brain Res. 2016;314:165–172. doi: 10.1016/j.bbr.2016.08.015. [DOI] [PubMed] [Google Scholar]
- 61.Zhao H, Mitchell S, Koumpa S, et al. Heme oxygenase-1 mediates neuroprotection conferred by argon in combination with hypothermia in neonatal hypoxia-ischemia brain injury. Anesthesiology. 2016;125:180–192. doi: 10.1097/ALN.0000000000001128. [DOI] [PubMed] [Google Scholar]
- 62.Höllig A, Weinandy A, Liu J, Clusmann H, Rossaint R, Coburn M. Beneficial properties of argon after experimental subarachnoid hemorrhage: early treatment reduces mortality and influences hippocampal protein expression. Crit Care Med. 2016;44:e520–529. doi: 10.1097/CCM.0000000000001561. [DOI] [PubMed] [Google Scholar]
- 63.Lavaur J, Le Nogue D, Lemaire M, et al. The noble gas xenon provides protection and trophic stimulation to midbrain dopamine neurons. J Neurochem. 2017;142:14–28. doi: 10.1111/jnc.14041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lavaur J, Lemaire M, Pype J, Le Nogue D, Hirsch EC, Michel PP. Xenon-mediated neuroprotection in response to sustained, low-level excitotoxic stress. Cell Death Discov. 2016;2:16018. doi: 10.1038/cddiscovery.2016.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Harris K, Armstrong SP, Campos-Pires R, Kiru L, Franks NP, 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. doi: 10.1097/ALN.0b013e3182a2a265. [DOI] [PubMed] [Google Scholar]
- 66.Veldeman M, Coburn M, Rossaint R, et al. 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. doi: 10.3389/fneur.2017.00511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Grim PS, Gottlieb LJ, Boddie A, Batson E. Hyperbaric oxygen therapy. JAMA. 1990;263:2216–2220. [PubMed] [Google Scholar]
- 68.Moon RE. Hyperbaric oxygen treatment for decompression sickness. Undersea Hyperb Med. 2014;41:151–157. [PubMed] [Google Scholar]
- 69.Kajiya M, Silva MJ, Sato K, Ouhara K, Kawai T. Hydrogen mediates suppression of colon inflammation induced by dextran sodium sulfate. Biochem Biophys Res Commun. 2009;386:11–15. doi: 10.1016/j.bbrc.2009.05.117. [DOI] [PubMed] [Google Scholar]
- 70.Vadas D, Kalichman L, Hadanny A, Efrati S. Hyperbaric oxygen environment can enhance brain activity and multitasking performance. Front Integr Neurosci. 2017;11:25. doi: 10.3389/fnint.2017.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dole M, Wilson FR, Fife WP. Hyperbaric hydrogen therapy: a possible treatment for cancer. Science. 1975;190:152–154. doi: 10.1126/science.1166304. [DOI] [PubMed] [Google Scholar]
- 72.Zhang Y, Sun Q, He B, Xiao J, Wang Z, Sun X. Anti-inflammatory effect of hydrogen-rich saline in a rat model of regional myocardial ischemia and reperfusion. Int J Cardiol. 2011;148:91–95. doi: 10.1016/j.ijcard.2010.08.058. [DOI] [PubMed] [Google Scholar]
- 73.Chen C, Chen Q, Mao Y, et al. Hydrogen-rich saline protects against spinal cord injury in rats. Neurochem Res. 2010;35:1111–1118. doi: 10.1007/s11064-010-0162-y. [DOI] [PubMed] [Google Scholar]
- 74.Nemeth J, Toth-Szuki V, Varga V, Kovacs V, Remzso G, Domoki F. Molecular hydrogen affords neuroprotection in a translational piglet model of hypoxic-ischemic encephalopathy. J Physiol Pharmacol. 2016;67:677–689. [PubMed] [Google Scholar]
- 75.Han L, Tian R, Yan H, et al. Hydrogen-rich water protects against ischemic brain injury in rats by regulating calcium buffering proteins. Brain Res. 2015;1615:129–138. doi: 10.1016/j.brainres.2015.04.038. [DOI] [PubMed] [Google Scholar]
- 76.Takeuchi S, Nagatani K, Otani N, Wada K, Mori K. Hydrogen does not Exert Neuroprotective Effects or Improve Functional Outcomes in Rats After Intracerebral Hemorrhage. Turk Neurosurg. 2016;26:854–859. doi: 10.5137/1019-5149.JTN.14098-15.1. [DOI] [PubMed] [Google Scholar]
- 77.Friis ML, Paulson OB, Hertz MM. Carbon dioxide permeability of the blood-brain barrier in man. The effect of acetazolamide. Microvasc Res. 1980;20:71–80. doi: 10.1016/0026-2862(80)90020-5. [DOI] [PubMed] [Google Scholar]
- 78.Vannucci RC, Brucklacher RM, Vannucci SJ. Effect of carbon dioxide on cerebral metabolism during hypoxia-ischemia in the immature rat. Pediatr Res. 1997;42:24–29. doi: 10.1203/00006450-199707000-00005. [DOI] [PubMed] [Google Scholar]
- 79.Ni Chonghaile M, Higgins BD, Costello JF, Laffey JG. Hypercapnic acidosis attenuates severe acute bacterial pneumonia-induced lung injury by a neutrophil-independent mechanism. Crit Care Med. 2008;36:3135–3144. doi: 10.1097/CCM.0b013e31818f0d13. [DOI] [PubMed] [Google Scholar]
- 80.Chonghaile MN, Higgins BD, Costello J, Laffey JG. Hypercapnic acidosis attenuates lung injury induced by established bacterial pneumonia. Anesthesiology. 2008;109:837–848. doi: 10.1097/ALN.0b013e3181895fb7. [DOI] [PubMed] [Google Scholar]
- 81.Westermaier T, Stetter C, Kunze E, et al. Controlled transient hypercapnia: a novel approach for the treatment of delayed cerebral ischemia after subarachnoid hemorrhage? J Neurosurg. 2014;121:1056–1062. doi: 10.3171/2014.7.JNS132611. [DOI] [PubMed] [Google Scholar]
- 82.Paul BD, Snyder SH. Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. Biochem Pharmacol. 2018;149:101–109. doi: 10.1016/j.bcp.2017.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Panthi S, Chung HJ, Jung J, Jeong NY. Physiological importance of hydrogen sulfide: emerging potent neuroprotector and neuromodulator. Oxid Med Cell Longev. 2016;2016:9049782. doi: 10.1155/2016/9049782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kimura H. Production and physiological effects of hydrogen sulfide. Antioxid Redox Signal. 2014;20:783–793. doi: 10.1089/ars.2013.5309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Karimi SA, Hosseinmardi N, Janahmadi M, Sayyah M, Hajisoltani R. The protective effect of hydrogen sulfide (H2S) on traumatic brain injury (TBI) induced memory deficits in rats. Brain Res Bull. 2017;134:177–182. doi: 10.1016/j.brainresbull.2017.07.014. [DOI] [PubMed] [Google Scholar]
- 86.Wink DA, Hanbauer I, Grisham MB, et al. Chemical biology of nitric oxide: regulation and protective and toxic mechanisms. Curr Top Cell Regul. 1996;34:159–187. doi: 10.1016/s0070-2137(96)80006-9. [DOI] [PubMed] [Google Scholar]
- 87.Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature. 1987;327:524–526. doi: 10.1038/327524a0. [DOI] [PubMed] [Google Scholar]
- 88.Beck-Schimmer B, Baumann L, Restin T, et al. Sevoflurane attenuates systemic inflammation compared with propofol, but does not modulate neuro-inflammation: A laboratory rat study. Eur J Anaesthesiol. 2017;34:764–775. doi: 10.1097/EJA.0000000000000668. [DOI] [PubMed] [Google Scholar]
- 89.Gaidhani N, Sun F, Schreihofer D, Uteshev VV. Duration of isoflurane-based surgical anesthesia determines severity of brain injury and neurological deficits after a transient focal ischemia in young adult rats. Brain Res Bull. 2017;134:168–176. doi: 10.1016/j.brainresbull.2017.07.018. [DOI] [PubMed] [Google Scholar]
- 90.Chen X, Zhou X, Yang L, et al. Neonatal exposure to low-dose (1.2%) sevoflurane increases rats' hippocampal neurogenesis and synaptic plasticity in later life. Neurotox Res. 2018;34:188–197. doi: 10.1007/s12640-018-9877-3. [DOI] [PubMed] [Google Scholar]
- 91.Zhou X, Li W, Chen X, et al. Dose-dependent effects of sevoflurane exposure during early lifetime on apoptosis in hippocampus and neurocognitive outcomes in Sprague-Dawley rats. Int J Physiol Pathophysiol Pharmacol. 2016;8:111–119. [PMC free article] [PubMed] [Google Scholar]
- 92.Xia Y, Xu H, Jia C, et al. Tanshinone IIA attenuates sevoflurane neurotoxicity in neonatal mice. Anesth Analg. 2017;124:1244–1252. doi: 10.1213/ANE.0000000000001942. [DOI] [PubMed] [Google Scholar]
- 93.Edwards DA, Shah HP, Cao W, Gravenstein N, Seubert CN, Martynyuk AE. Bumetanide alleviates epileptogenic and neurotoxic effects of sevoflurane in neonatal rat brain. Anesthesiology. 2010;112:567–575. doi: 10.1097/ALN.0b013e3181cf9138. [DOI] [PubMed] [Google Scholar]
- 94.Baddeley AD, Flemming NC. The efficiency of divers breathing oxy-helium. Ergonomics. 1967;10:311–319. doi: 10.1080/00140136708930873. [DOI] [PubMed] [Google Scholar]
- 95.Aehling C, Weber NC, Zuurbier CJ, et al. Effects of combined helium pre/post-conditioning on the brain and heart in a rat resuscitation model. Acta Anaesthesiol Scand. 2018;62:63–74. doi: 10.1111/aas.13041. [DOI] [PubMed] [Google Scholar]
- 96.Gardner AJ, Menon DK. 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. [DOI] [PubMed] [Google Scholar]
- 97.Amer AR, Oorschot DE. Xenon combined with hypothermia in perinatal hypoxic-ischemic encephalopathy: a noble gas, a Noble mission. Pediatr Neurol. 2018;84:5–10. doi: 10.1016/j.pediatrneurol.2018.02.009. [DOI] [PubMed] [Google Scholar]