Back to Journals » Clinical Interventions in Aging » Volume 20

Normobaric Hyperoxia Therapy in Treating Stroke

Authors Duan Y, Li X, Han S, Guan J, Chen Z, Li W, Meng R , Ding J 

Received 9 February 2025

Accepted for publication 27 June 2025

Published 2 July 2025 Volume 2025:20 Pages 969—981

DOI https://doi.org/10.2147/CIA.S521584

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Maddalena Illario



Yanli Duan,1,* Xiangyu Li,2,* Song Han,1 Jingwei Guan,1 Zhiying Chen,3 Weili Li,4 Ran Meng,5 Jiayue Ding1

1Department of Neurology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China; 2Department of Neurology, Tianjin Huanhu Hospital, Tianjin, People’s Republic of China; 3Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, People’s Republic of China; 4Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, People’s Republic of China; 5Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Jiayue Ding, Department of Neurology, Tianjin Medical University General Hospital, No. 154 Anshan Street, Heping District, Tianjin, 300052, People’s Republic of China, Email [email protected]

Abstract: Normobaric hyperoxia (NBO) is a standard oxygenation intervention for various conditions/diseases including stroke. The present review summarizes the current literature addressing the neuroprotective mechanisms of NBO in acute ischemic stroke (AIS), intracranial hemorrhage, and chronic cerebral ischemia, as well as its combination with other therapies to identify a more appropriate and effective NBO treatment method and to benefit more patients in clinical settings. The primary mechanism of action of NBO is the elevation of the interstitial partial pressure of oxygen in arterial blood (PaO2) in brain tissue. NBO preconditioning yields moderate production of free radicals before AIS, which can increase antioxidant enzyme production, alter mitochondrial membrane lipids, increase tumor necrosis factor-alpha (TNF-α) converting enzyme levels, stimulate the hypoxia-inducible factor signaling pathway, upregulate glutamate transporters, Na+–Ca+ exchanger, and the metabotropic glutamate receptor after AIS, thus conferring neuroprotection to brain tissue. NBO postconditioning benefits AIS by protecting the penumbra and extending the recanalization time window, indicating that reperfusion is critical for the beneficial effects of NBO. Some previous clinical trials have obtained negative results because they enrolled non-reperfused cohorts. Given penumbra protection, NBO can enhance the efficacy of recanalization therapy, including thrombolysis and endovascular treatment. Clinical studies have indicated that NBO benefits only patients with reperfusion, which is consistent with animal-based research. NBO combined with medications, such as ethanol, minocycline, and edaravone, can more effectively treat AIS than NBO alone. Moreover, NBO demonstrates promise for the treatment of intracranial hemorrhage and chronic cerebral ischemia. NBO is a safe and effective therapy for stroke; however, eligible populations should be restricted to those with penumbra or ischemic and hypoxic brain tissues.

Keywords: normobaric hyperoxia, ischemic stroke, hemorrhagic stroke, penumbra

Introduction

Normobaric hyperoxia (NBO), supplied via cannula or facemask (eg, simple mask) at 1 atmosphere pressure (101.325 kPa), is a standard oxygenation intervention used for various conditions/diseases, particularly stroke.1–3 Oxygen can easily diffuse across the blood-brain barrier (BBB) to rapidly increase oxygen concentration in brain tissue.4 Although long-term oxygen supplementation is harmful to organs, short durations or intermittent use is safe, well tolerated, and effective in various populations.5,6 In those diagnosed with acute ischemic stroke (AIS), the primary goal of NBO is to protect the penumbra from infarction. It is well-established that NBO can significantly increase the interstitial partial pressure of oxygen in arterial blood (PaO2) close to the pre-ischemic level in the ischemic penumbra, but not in the core.7,8 In theory, high oxygen concentration(s) contribute to oxidative stress-related injury; however, oxygen therapy does not appreciably increase oxygen radical generation and damage in cerebral ischemia.9 Regarding clinical settings, a large body of retrospective research indicates that increasing the oxygenation index in patients diagnosed with AIS is associated with lower in-hospital mortality.10 The advantages of NBO in treating AIS are clear. Based on the protective efficacy of NBO on the penumbra in AIS, NBO may not only mitigate hypoxic damage in perihematoma in intracranial hemorrhage (ICH), but also reduce chronic injury in ischemic hypoxic brain tissue in chronic cerebral ischemia (CCI).11,12 Moreover, given the mechanisms of NBO in treating ischemic stroke, recanalization or reflow within a short period is critical to the protective process. As such, selecting the most appropriate methods and suitable population(s) is very important for effective NBO treatment of stroke. Accordingly, the present review summarizes the current literature addressing the neuroprotective mechanisms of preconditioning and postconditioning NBO in stroke (AIS, ICH, and CCI), as well as its combination with other therapies, to identify a more appropriate and effective NBO treatment method and to benefit more patients in clinical settings.

The Time Course of Normobaric Hyperoxia on Acute Ischemic Stroke

Preconditioning with Normobaric Hyperoxia on Acute Ischemic Stroke

Ischemic tolerance is an important mechanism for protecting the brain tissue from severe ischemia. NBO preconditioning can also relieve ischemic injury in the brain tissue. Extensive research by Bigdeli et al demonstrated that pre-ischemic intermittent NBO—rather than prolonged exposure to hyperoxia—can simulate the moderate production of oxygen-free and hydroxyl radicals.13 Repeated sublethal injuries increases antioxidant enzyme production (superoxide dismutase, catalase, glutathione peroxidase, peroxidase, and glutathione reductase), which play a pivotal role in brain ischemic tolerance.13–15 Moreover, during both ischemia and reperfusion (I/R), a large amount of free radicals is produced in the brain, leading to incremental changes in mitochondrial membrane lipids. The elevation of mitochondrial membrane lipids causes mitochondrial decay, failure of the electron transmission chain, and inhibition of energy production, ultimately leading to cell death.16 NBO preconditioning induces changes in the brain lipidome, including cholesterol ester, triglyceride, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylcholine, ceramide, cerebroside, cholesterol and sphingomyelin, all of which prevent free radical-induced injury.17

Preconditioning with NBO upregulates glutamate transporters (EAAT1, EAAT2 and EAAT3) in the rat brain.18,19 Glutamate transporters in neurons and glia remove glutamate from the extracellular space, preventing the concentration of extracellular glutamate from increasing to neurotoxic levels.20,21 Inhibition of the glial glutamate transporter EAAT2 aggravates brain edema after transient focal cerebral ischemia in rats.19 Glutamate is highly expressed in the core and penumbra of cerebral infarction, where it over-activates ionotropic glutamate receptors (NMDA and AMPA) causing an influx of calcium (Ca2+) and sodium (Na+) ions. This process facilitates protease activation, lipase activation, free-radical increase, altered transcription, protein kinase C activation, hydrolysis of protein phosphatase inhibitor, and cell swelling, ultimately leading to subsequent cell death.22 Therefore, the elevation of glutamate transporters induced by NBO preconditioning can prevent brain tissue from glutamate-induced injury. Simultaneously, NBO preconditioning can also upregulate the Na+–Ca+ exchanger (NCX family, NCX1, NCX2 and NCX3), which can reduce the extent of brain infarct volume after brain ischemia.23,24 NBO preconditioning has another effect on upregulating both metabotropic glutamate receptor (mGluR) I and II.25 mGluR I and II are associated with oxidative stress in the brain.26 Increased mGluR I and II lead to an increment in glutathione levels that exert neuroprotection on the ischemic brain after intermittent NBO treatment.25 Furthermore, NBO preconditioning increases tumor necrosis factor-alpha (TNF-α) converting enzyme (TACE) expression, consistent with a protective role for TNF-α release.18,19 TACE expression contributes to a rise in TNF-α, which participates in the development of ischemic tolerance after excitotoxic stimuli.27,28 As mentioned, NBO-induced upregulation of glutamate transporters may, in part, be mediated by the TACE/TNF-α pathway.18,29 NBO preconditioning also stimulates the hypoxia-inducible factor (HIF) signaling pathway, causing increased expression of vascular endothelial growth factor (VEGF) and erythropoietin.30 The HIF signaling pathway regulates neurogenesis and angiogenesis through increased VEGF and erythropoietin, leading to neural stem cell proliferation and migration of neural progenitors.31,32

The interval between the initiation of preconditioning NBO therapy and the onset of ischemic stroke significantly affects the neuroprotective effects conferred by NBO. Specifically, ischemia occurring 15 days after the completion of intermittent NBO preconditioning is associated with higher Neurological Deficit Score, larger infarct volumes, and increased cerebral edema compared with ischemia occurring merely 2 days post-treatment.33 These findings suggest that, while NBO preconditioning induces effective neuroprotection, its benefits are transient, potentially due to a gradual decline in the antioxidant response and ischemic tolerance induced by preconditioning therapy. In summary, the primary mechanistic goals of NBO preconditioning in the treatment of AIS are to decrease oxidative stress-related injury, reduce glutamate-induced damage, and stimulate neurogenesis and angiogenesis after an ischemic attack (Figure 1). NBO therapy may represent a promising approach for both primary and secondary prevention of ischemic stroke. However, clinical trials remain scarce despite a substantial body of animal-based research supporting the effectiveness of pre-ischemic NBO treatment.

Figure 1 Mechanisms of normobaric hyperoxia (NBO) therapy in acute ischemic stroke (AIS). NBO preconditioning increases antioxidant enzyme production, alters mitochondrial membrane lipids, increases tumor necrosis factor-alpha (TNF-α) converting enzyme levels, stimulates the hypoxia-inducible factor signaling pathway, and upregulate glutamate transporters, Na+–Ca+ exchanger, and metabotropic glutamate receptor after AIS. NBO postconditioning can reduce infarction volume, improve metabolism, increase tissue perfusion, reduce free-radical generation, ameliorate hyperglycolytic cascades, maintain mitochondrial function, and preserve blood-brain barrier integrity. Both NBO preconditioning and postconditioning reduce infarction volume, ameliorate brain edema, and improve neurological deficits.

Effect of Normobaric Hyperoxia Postconditioning on Acute Ischemic Stroke

NBO is routinely performed after stroke in clinical settings. Therefore, animal studies investigating the effects of post-ischemic NBO are considered to be more relevant for guiding clinical treatment. It is widely recognized that NBO yields significant efficacy in reducing infarct volume, especially in ischemic models with reperfusion.34–37 In other words, the primary neuroprotection conferred by NBO is to extend the reperfusion window and offer lasting tissue protection in temporary ischemia.38 Kim et al indicated that NBO widened the reperfusion time window in rats by up to 2 h without increasing superoxide radical generation. However, the benefit of NBO is not sustained in permanent ischemia.39,40 Thus, NBO may be the most appropriate adjunct therapy to extend the time window for recanalization treatment (thrombolysis and endovascular treatment [EVT]), rather than routine medical treatment.39

Using magnetic resonance imaging (MRI), Singhal et al demonstrated that NBO can enhance aerobic metabolism, preserve neuronal integrity and reduce MRI abnormalities in the ischemic brain.41–44 Given the efficacy in increasing interstitial PaO2 in the penumbra, other studies have shown that NBO significantly reduces ADP/ATP ratio, reactive oxygen species (ROS) levels, and tissue acidosis after AIS, indicating restoration of impaired energy metabolism.45,46 Moreover, the expression of glucose transporters 1 and 3 (Glut1 and 3) and the glycolytic rate-limiting enzyme phosphofructokinase-1 (PFK-1), which results in hyperglycolytic cascade, are attenuated by NBO via AMPK signaling.45

In addition to elevating interstitial PaO2 in the penumbra, NBO can also augment or preserve tissue perfusion.8,36,41,42,44,47,48 The mechanism of improved ischemic tissue perfusion by NBO may be attributed to enhanced nitric oxide (NO) production by endothelial NO synthase (eNOS) through O2 substrate availability, suppression of peri-infarct depolarization, and vasoconstrictive effects on circulation in the penumbra.47,48

NADPH oxidase is known to produce ROS after cerebral I/R.49 The NADPH catalytic subunit gp91phox (or Nox2) is critically implicated in AIS and knockout of Nox2 results in significant reduction of NADPH oxidase activity during ischemia.50,51 It has been demonstrated that NBO can attenuate Nox2-containing NADPH oxidase, which is an important mechanism underlying NBO-afforded brain protection.51–55 Through inhibiting NADPH oxidase, NBO treatment can relieve oxidative stress-related injury, suppress matrix metalloproteinase-9 (MMP-9) induction and inhibit the degradation of occludin, attenuating of disruption of the BBB and the development of edema in the brain.8,51,53,56–58 Under ischemic conditions, several proposed mechanisms are associated with the activation of Nox2, such as phosphatidylinositol 3-kinase (PI3K)/Akt pathways, inflammatory cytokines and metabotropic glutamate receptors.59–61 NBO treatment reduces reperfusion injury through activation of the PI3K/Akt pathway.62 In addition to oxygen radicals, other free radicals, such as reactive nitrogen species—formed by the reaction of NO with superoxide under ischemic conditions—is also alleviated by NBO via suppressing neuronal NO synthase.63 By retarding free-radical generation, NBO can relieve ischemic damage, micro-circulation failure, and tissue inflammation.

NBO also has a beneficial effect on mitochondria. NBO alleviates ischemia-induced cytochrome c (CytC) release from mitochondria through stimulating the GPX4-connexin43 pathway in astrocytes.64 As such, NBO can reduce mitochondrial damage by maintaining redox homeostasis in astrocytes, rather than increasing oxidative stress.64 Furthermore, NBO can also reduce cytosolic free zinc accumulation in the penumbra and suppress zinc overload in mitochondria after ischemic stroke, which improve mitochondrial membrane potential and, ultimately, protect neurons in the penumbra.65 Mitochondrial integrity maintains cell survival. Therefore, NBO attenuates autophagy, which is overactivated by I/R.66 Meanwhile, NBO decreases caspase-8 cleavage in the penumbra during I/R.8

The membrane proteins Na+/H+ exchanger 1 (NHE1) and aquaporin 4 (AQP4) play a pivotal role in the development of brain edema after ischemia.67 NHE1 is associated with H+ extrusion in exchange for Na+ in astrocytes, which maintain BBB function.68 Excessive NHE1 activation during ischemia is crucial for Na+ overload, resulting in Ca2+ entry via reversal of the Na+/Ca2+ exchanger (NCX).69 The increased cytosolic Ca2+ triggers a neurotoxic cascade.68,69 Similarly, AQP4, which is mainly expressed in perivascular astrocyte end-feet, is also associated with BBB function.70 Increased AQP4 level(s) can destroy BBB integrity, leading to water entering brain tissues, resulting in brain edema.71 NBO is capable of retarding exacerbation of brain edema through suppressing the expression of NHE1 and AQP4.72

Similar to NBO preconditioning, the timing and method of NBO post-conditioning also affect neuroprotection. Compared with NBO initiated during ischemia, NBO delivered during reperfusion is unlikely to have a protective impact and may even exacerbate neuronal injury and death in brain tissue, despite reducing excitotoxic calcium influx and subsequent degeneration.8,73 Liu et al reported that intermittent NBO (oxygen oscillation) during ischemia yielded more neuroprotection than continuous or short NBO.62 Therefore, intermittent NBO starting during ischemia is critical to maximize its benefit for patients diagnosed with AIS. The mechanisms of NBO postconditioning are described in Figure 1.

The efficacy of NBO in cerebral ischemia has been tested in multiple clinical trials; however, the results have been disappointing (Table 1).6,42,43,58,74–85 We and other teams have analyzed the existing literature, and the secondary analysis revealed limited evidence supporting NBO for the treatment of stroke.86,87 Studies investigating the effect of NBO on AIS imaging tests have reached beneficial conclusions; however, those investigating the effect of NBO on the clinical neurological and functional scores of patients without recanalization therapies have reached negative—or even harmful—conclusions. In addition, studies enrolling patients undergoing recanalization therapies, including thrombolysis and EVT, especially those diagnosed with anterior circulation AIS, have demonstrated the beneficial effects of NBO. Confirmation of the null hypothesis has been due to most enrolled patients not achieving reperfusion during NBO. As discussed, the primary mechanistic goal of NBO is to “freeze the penumbra”; therefore, without blood retention, NBO cannot inhibit infarction expansion. Imaging tests assess perfusion and metabolism, revealing the effects of NBO on the brain tissue during the acute phase of AIS. Without reperfusion after the acute phase, the penumbra progresses to an infarction, thus eliminating the neuroprotective effects of NBO. When reperfusion occurs through recanalization therapies, the penumbra protected by NBO changes to normal brain tissue, thereby enhancing the neuroprotective effect of NBO. Therefore, reperfusion therapy alone may enhance the efficacy of NBO in cerebral ischemia.

Table 1 The Efficacy of NBO in Clinical Studies

Combining Normobaric Hyperoxia with Other Adjuvant Therapies in Acute Ischemic Stroke

Adjuvant Use of Normobaric Hyperoxia in Intravenous Thrombolysis

Considering the neuroprotective role of NBO in the ischemic penumbra and BBB, it is considered to be an ideal adjunct therapy to improve the efficacy and safety of tissue-type plasminogen activators (tPA). The most important effect of NBO on ischemia is the expansion of the therapeutic time window for successful reperfusion during tPA treatment, especially under delayed tPA administration. Animal studies have demonstrated that early NBO treatment could slow the evolution of ischemic BBB damage, decrease MMP-9 production, and reduce the loss of tight junction proteins (TJPs), leading to significant neurological improvement and reduced infarction volume, brain edema, cerebral hemorrhage and mortality after delayed tPA treatment.54,89–91 Imaging studies have revealed that 60 min NBO treatment before thrombolysis could reduce infarct size and decrease the frequency of gross parenchymal hemorrhage by reducing early BBB permeability and improving microvascular integrity after tPA-induced reperfusion.92 In clinical settings, a retrospective study found that NBO combined with intravenous thrombolysis could reduce infarct volume and improve long-term outcomes.83 However, a prospective or randomized controlled trial is lacking. The OPEN-3 clinical trial is an ongoing multicenter, randomized, controlled Phase III study in China that aims to evaluate the safety and efficacy of NBO in patients undergoing intravenous thrombolysis (NCT05965687). We anticipate that these results will further consolidate evidence supporting the effect of NBO on ischemic stroke.

However, other studies have reported conflicting results. Fujiwara et al did not find a beneficial effect of NBO on augmenting the efficacy of tPA thrombolysis and noted no induced cerebral hemorrhage or edema after NBO.93 David et al indicated that combining NBO and tPA exacerbated ischemic brain damage and swelling compared with NBO or tPA alone, because NBO facilitated the catalytic and thrombolytic efficiency of tPA; the synergistic effect of combined NBO and tPA may worsen brain injury.94 Notably, NBO combined with tPA performed after reperfusion could induce oxidative stress-related injury, particularly in cases with severe infarction, which may lead to brain edema and hemorrhagic transformation. Therefore, NBO should be initiated during the ischemic phase regardless of tPA administration.

Adjuvant Use of Normobaric Hyperoxia in Endovascular Treatment

Because the penumbra can potentially be rescued if blood flow is restored in a timely manner, NBO may serve as an early intervention to preserve the penumbra and “buy time” for combination therapies to achieve better outcomes.38,95 As mentioned, whether early or late use of NBO, if cessation of blood supply is relieved, NBO can reduce the final infarction volume. Therefore, patients with AIS receiving rapid blood flow restoration through EVT appear to be more suitable for NBO treatment in clinical settings. Recent randomized controlled trials revealed that NBO therapy (100% O2 via a face mask, 10 L/min) for 4 h in patients who underwent EVT, can significantly reduce the infarction volume within 24–48 h and result in lower modified Rankin Scale (mRS) scores at 90 days. In addition, the safety of NBO is also satisfactory, as the incidences of ICH, death, and other adverse events were similar to those without NBO.74,88 Serum assessments find that NBO can maintain BBB integrity and reduce brain injury in patients with AIS undergoing EVT.96 Further dose-escalation study demonstrated that NBO therapy for 4 and 6 hours appeared to be more effective.97 Another earlier randomized controlled trial reached a similar conclusion using a different NBO protocol (50% O2 via a face mask, 15 L/min) started after vessel recanalization and continued for 6 h. The trial revealed that NBO could improve the degree of 90-day disability, decrease 90-day mortality, and profoundly reduce 24 h infarction volume, without increasing adverse events.84 Although these trials obtained a favorable conclusion, it is noteworthy that they only enrolled patients with anterior circulation stroke, excluding those with posterior circulation stroke. Only 1 randomized trial currently focuses on NBO after EVT for posterior circulation stroke; however, despite using the same NBO treatment paradigm, it only found a trend, rather than a significant effect of NBO in this cohort.85 The reason for the different effectiveness of NBO on anterior and posterior circulation stroke after EVT remains unknown. We considered that this may be because the brainstem has a smaller penumbra than the hemisphere or basal ganglia due to the smaller infarction volume in posterior circulation stroke. The small penumbra limits salvage of the ischemic brain tissue and further suppresses the efficacy of NBO. Therefore, we believe that NBO is an appropriate treatment strategy for patients undergoing EVT, especially those with anterior circulation stroke.

Nevertheless, opposing viewpoints exist. The primary mechanism of action of NBO treatment for AIS involves elevated PaO2. In other words, a high PaO2 appears to confer beneficial neuroprotective effects against AIS. However, a prospective observational study found that patients with admission PaO2 > 120 mmHg experienced worse functional outcomes and higher mortality at 90 days after EVT compared to those with admission PaO2 ≤ 120 mmHg.98 It should be noted that the oxygen therapy in this study was in the form of mechanical ventilation, which is completely different from NBO. Additionally, this was an observational study that included all consecutively admitted patients, rather than a randomized grouping and intervention. These 2 points could have biased the results toward the null hypothesis.

Combining Normobaric Hyperoxia with Medication

Several studies have investigated whether other adjuvant therapies can enhance the efficacy of NBO in AIS. Ethanol therapy is a candidate for combination therapy.99 Cai et al found that combined NBO and ethanol therapy modulated pyruvate dehydrogenase, demonstrated by upregulated pyruvate dehydrogenase complex (PDH) and downregulated pyruvate dehydrogenase kinase (PDK).100 The combined therapy also decreased lactic acidosis, Glut1, Glut3, PFK-1 and lactate dehydrogenase (LDH), indicating hyperglycolysis was suppressed.101 In addition, the combination therapy inhibits PKC-Akt-NOX pathway and reduces cerebral monocarboxylate transporters.102,103 All of the aforementioned mechanisms stabilize anaerobic metabolism, decrease oxidative damage and retard cell apoptosis, thus improving neurological deficit and reducing infarct volume.91,100–106

Minocycline is another treatment that combines NBO and ischemic stroke. Jin et al found that, compared with NBO or minocycline alone, combination treatment significantly reduced infarction volume and hemispheric swelling by reducing the levels of MMP-2, MMP-9, caspase-9 and caspase-3.107 Other adjuvant drugs, such as N-acetylcysteine, edaravone, cilostazol, melatonin, and methylene blue, have been shown to reduce infarction volume, protect BBB integrity, and improve neurological deficits; however, the supportive evidence is weak.108–112

Normobaric Hyperoxia Used in the Treatment of Intracranial Hemorrhage

Theoretically, ICH-induced mechanical compression of the surrounding brain tissue is a major injury that can decrease aerobic metabolism and perfusion in the perihematoma, causing edema and even enlarging the hematoma. Similar to the ischemic penumbra, low perfusion and abnormal metabolism in the perihematoma exposes brain tissue to an ischemic hypoxic condition. Therefore, rescue of the perihematoma is key for treating ICH for which NBO may be a promising therapeutic candidate.113 Zhou et al demonstrated that initiating NBO 30 min after inducing ICH significantly reduced hemorrhage volume, mitigated the development of post-hemorrhagic edema, and preserved BBB integrity, whereas NBO initiated at 60 min or 120 min yielded no beneficial effects.114,115 NBO appears to attenuate the post-hemorrhagic breakdown of occludin, inhibit the activation of MMP-9 and suppress the upregulation of HIF-1α, collectively contributing to the reduction of hemorrhage volume and stabilization of the BBB. Similar to the ischemic puncture, perihematomal tissue exhibits a critical therapeutic window. Delayed NBO therapy may be effective or even detrimental because progressive injury to the perihematomal region and NBO-induced increases in intracellular oxygen levels in damaged cells can enhance ROS production, thereby exerting deleterious effects. Furthermore, given that MMPs and HIF-1α are involved in acute pathophysiological responses and extracellular matrix remodeling, as well as angiogenesis and neurogenesis during the delayed phase of brain injury, delayed inhibition of these pathways may evoke deleterious effects.114,116,117 In addition, NBO also suppresses VEGF expression, which may inhibit cellular apoptosis potentially through the VEGF/PI3K/Akt/FoxO1 pathway.118,119 NBO can effectively decrease the level of C3 which is able to rescuing microglia-mediated synaptic pruning after ICH.120 There is no published clinical trial; however, a single-center, randomized controlled Phase II trial from our team registered a clinical trial (Clinicaltrials.gov, NCT04144868) that revealed the beneficial effect of NBO on ICH in clinical settings. Our findings indicate that early and intermittent administration of NBO in patients with ICH significantly improves 90-day prognosis, likely due to a reduction in perihematoma edema induced by NBO therapy. A small sample size, however, renders the study underpowered and restricts its clinical scope. As such, a well-designed, multicenter, randomized, controlled phase III trial is warranted to draw more convincing conclusions. NBO appears to be a promising approach to treating ICH, and more studies should be conducted to verify this conclusion.

Normobaric Hyperoxia Used in the Treatment of Chronic Cerebral Ischemia

CCI is a pathological condition induced by a persistent reduction in cerebral perfusion, leading to ischemia and hypoxia in brain tissue. CCI can cause various brain dysfunctions such as dizziness, cognitive impairment, and stroke.121,122 Cerebrovascular atherosclerotic stenosis is the most common pathogenesis of CCI, with hypoperfusion being a pivotal mechanism accounting for these neurological deficits.122,123 In theory, low cerebral blood flow in patients with CCI exposes the brain tissue to an ischemic-hypoxic condition, which is similar to that in the ischemic penumbra in brain infarction.121 Therefore, given the satisfactory effectiveness of NBO on the penumbra in AIS, NBO may also yield beneficial effects on the ischemic-hypoxic brain tissue. Based on previous animal and clinical studies focusing on NBO in AIS, we hypothesize that NBO may also be a novel approach to the treatment of CCI.12 To verify this, we evaluated the improvement of brain dysfunction in patients with CCI treated with NBO. Electroencephalography revealed that NBO rapidly ameliorated CCI-mediated brain wave anomalies, including reduced abnormal high-power oscillations and slow paroxysmal activity associated with CCI, which may improve vascular cognitive impairment.124

Aside from cerebral arterial stenosis-mediated CCI, a form of CCI associated with cerebral venous stenosis has recently been proposed.125–127 Venous outflow disturbance raises the pressure in arterio-venous anastomoses, thereby affecting arterial blood flow and volume.128,129 Consequently, cerebral venous outflow disturbance may result in global brain hypoperfusion and elevated aerobic metabolism, a pattern similar to that observed in arterial stenosis-related CCI.126 NBO therapy can elevate cerebral tissue pO2 and may ameliorate cerebral venous outflow disturbance-related deficits, especially headache and electroencephalogram brain dysfunction.130 Current animal and clinical investigations of NBO for CCI are fewer than those for AIS; however, CCI populations are larger. Although we found preliminary beneficial effects of NBO, more studies are warranted to unravel mechanisms and determine effectiveness. CCI etiologies can be classified as large artery atherosclerotic stenosis, small arterial stenosis or occlusion, cardiogenic hypoperfusion, cerebral venous outflow disturbance, and other causes. Future NBO trials should stratify cohorts according to these subtypes.121

Conclusions

Although existing research remains at a relatively superficial level, we summarize the therapeutic mechanisms of NBO into 2 points. First, NBO elevates pO2 in the penumbra of AIS, as well as in perihemotoma of ICH and hypoxic-ischemic tissue of CCI, which reduces ischemia volume, improves metabolism, protects BBB integrity, and maintains mitochondrial function, thus expanding the therapeutic time window. Second, NBO increases the production of antioxidant enzymes and reduces free-radical generation, leading to alleviated oxygen stress-related injury in stroke. Reperfusion is critical for the beneficial effects of NBO in AIS. Therefore, only patients undergoing recanalization can benefit from NBO. Because aerobic metabolism plays a pivotal role in the NBO treatment of stroke, future research should emphasize the role of the mitochondria, particularly their involvement in mitophagy and fission/fusion dynamics. Given the heterogeneity observed in both animal and clinical studies, additional research is imperative to consolidate evidence supporting the effectiveness of NBO in treating stroke.

Acknowledgments

We would like to thank all of the patients and doctors who participated in this study for their cooperation.

Funding

This study was sponsored by the National Natural Science Foundation of China (82201432) and Tianjin Key Medical Discipline (Specialty) Construction Project.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Siemieniuk R, Chu DK, Kim LH, et al. Oxygen therapy for acutely ill medical patients: a clinical practice guideline. BMJ. 2018;363:k4169. doi:10.1136/bmj.k4169

2. Balestra C, Baldelli S, Virgili F, Salvagno M, Mrakic-Sposta S, Fratantonio D. Pulsed hyperoxia acts on plasmatic advanced glycation end products and advanced oxidation protein products and modulates mitochondrial biogenesis in human peripheral blood mononuclear cells: a pilot study on the “normobaric oxygen paradox”. Int J Mol Sci. 2024;25(4):2394. doi:10.3390/ijms25042394

3. Khalife M, Salvagno M, Sosnowski M, Balestra C. Exploring the effects of post operative hyperoxic intermittent stimuli on reticulocyte levels in cancer patients: a randomized controlled study. J Anesth Analg Crit Care. 2024;4(1):43. doi:10.1186/s44158-024-00179-x

4. Hawkins BT, Egleton RD. Pathophysiology of the blood-brain barrier: animal models and methods. Curr Top Dev Biol. 2008;80:277–309.

5. Li C, Jiang M, Chen Z, et al. The neuroprotective effects of normobaric oxygen therapy after stroke. CNS Neurosci Ther. 2024;30(7):e14858. doi:10.1111/cns.14858

6. Roffe C, Nevatte T, Sim J, et al. Effect of routine low-dose oxygen supplementation on death and disability in adults with acute stroke: the stroke oxygen study randomized clinical trial. JAMA. 2017;318(12):1125–1135. doi:10.1001/jama.2017.11463

7. Liu S, Shi H, Liu W, Furuichi T, Timmins GS, Liu KJ. Interstitial pO2 in ischemic penumbra and core are differentially affected following transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 2004;24(3):343–349. doi:10.1097/01.WCB.0000110047.43905.01

8. Liu S, Liu W, Ding W, Miyake M, Rosenberg GA, Liu KJ. Electron paramagnetic resonance-guided normobaric hyperoxia treatment protects the brain by maintaining penumbral oxygenation in a rat model of transient focal cerebral ischemia. J Cereb Blood Flow Metab. 2006;26(10):1274–1284. doi:10.1038/sj.jcbfm.9600277

9. Sun L, Wolferts G, Veltkamp R. Oxygen therapy does not increase production and damage induced by reactive oxygen species in focal cerebral ischemia. Neurosci Lett. 2014;577:1–5. doi:10.1016/j.neulet.2014.05.060

10. Akca O, Nichols J, Stewart B, Elliott C, Remmel K, Lenhardt R. Association of early oxygenation levels with mortality in acute ischemic stroke - a retrospective cohort study. J Stroke Cerebrovasc Dis. 2020;29(2):104556. doi:10.1016/j.jstrokecerebrovasdis.2019.104556

11. Zeng H, Zeng D, Yin X, Zhang W, Wu M, Chen Z. Research progress on high-concentration oxygen therapy after cerebral hemorrhage. Front Neurol. 2024;15:1410525. doi:10.3389/fneur.2024.1410525

12. Ding J, Zhou D, Liu C, et al. Normobaric oxygen: a novel approach for treating chronic cerebral circulation insufficiency. Clin Interv Aging. 2019;14:565–570. doi:10.2147/CIA.S190984

13. Bigdeli MR, Hajizadeh S, Froozandeh M, Rasulian B, Heidarianpour A, Khoshbaten A. Prolonged and intermittent normobaric hyperoxia induce different degrees of ischemic tolerance in rat brain tissue. Brain Res. 2007;1152:228–233. doi:10.1016/j.brainres.2007.03.068

14. Bigdeli MR, Rasoulian B, Meratan AA. In vivo normobaric hyperoxia preconditioning induces different degrees of antioxidant enzymes activities in rat brain tissue. Eur J Pharmacol. 2009;611(1–3):22–29. doi:10.1016/j.ejphar.2009.03.034

15. Bigdeli MR. Preconditioning with prolonged normobaric hyperoxia induces ischemic tolerance partly by upregulation of antioxidant enzymes in rat brain tissue. Brain Res. 2009;1260:47–54. doi:10.1016/j.brainres.2008.12.065

16. Bigdeli MR. Neuroprotection caused by hyperoxia preconditioning in animal stroke models. Sci World J. 2011;11:403–421. doi:10.1100/tsw.2011.23

17. Faezi M, Bigdeli MR, Mirzajani F, Ghasempour A. Normobaric hyperoxia preconditioning induces changes in the brain lipidome. Curr Neurovasc Res. 2015;12(4):353–362. doi:10.2174/1567202612666150819100746

18. Bigdeli MR, Hajizadeh S, Froozandeh M, et al. Normobaric hyperoxia induces ischemic tolerance and upregulation of glutamate transporters in the rat brain and serum TNF-alpha level. Exp Neurol. 2008;212(2):298–306. doi:10.1016/j.expneurol.2008.03.029

19. Bigdeli MR, Rahnema M, Khoshbaten A. Preconditioning with sublethal ischemia or intermittent normobaric hyperoxia up-regulates glutamate transporters and tumor necrosis factor-alpha converting enzyme in the rat brain. J Stroke Cerebrovasc Dis. 2009;18(5):336–342. doi:10.1016/j.jstrokecerebrovasdis.2008.12.005

20. Takahashi M, Billups B, Rossi D, Sarantis M, Hamann M, Attwell D. The role of glutamate transporters in glutamate homeostasis in the brain. J Exp Biol. 1997;200(Pt 2):401–409. doi:10.1242/jeb.200.2.401

21. Maragakis NJ, Rothstein JD. Glutamate transporters: animal models to neurologic disease. Neurobiol Dis. 2004;15(3):461–473. doi:10.1016/j.nbd.2003.12.007

22. Romera C, Hurtado O, Botella SH, et al. In vitro ischemic tolerance involves upregulation of glutamate transport partly mediated by the TACE/ADAM17-tumor necrosis factor-alpha pathway. J Neurosci. 2004;24(6):1350–1357. doi:10.1523/JNEUROSCI.1596-03.2004

23. Pignataro G, Tortiglione A, Scorziello A, et al. Evidence for a protective role played by the Na+/Ca2+ exchanger in cerebral ischemia induced by middle cerebral artery occlusion in male rats. Neuropharmacology. 2004;46(3):439–448. doi:10.1016/j.neuropharm.2003.09.015

24. Mohammadi E, Bigdeli MR. Effects of preconditioning with normobaric hyperoxia on Na+/Ca2+ exchanger in the rat brain. Neuroscience. 2013;237:277–284. doi:10.1016/j.neuroscience.2013.01.064

25. Nasrniya S, Bigdeli MR. Ischemic tolerance induced by normobaric hyperoxia and evaluation of group I and II metabotropic glutamate receptors. Curr Neurovasc Res. 2013;10(1):21–28. doi:10.2174/156720213804805981

26. Deng W, Wang H, Rosenberg PA, Volpe JJ, Jensen FE. Role of metabotropic glutamate receptors in oligodendrocyte excitotoxicity and oxidative stress. Proc Natl Acad Sci U S A. 2004;101(20):7751–7756. doi:10.1073/pnas.0307850101

27. Hurtado O, Cárdenas A, Lizasoain I, et al. Up-regulation of TNF-alpha convertase (TACE/ADAM17) after oxygen-glucose deprivation in rat forebrain slices. Neuropharmacology. 2001;40(8):1094–1102. doi:10.1016/S0028-3908(01)00035-1

28. Hurtado O, Lizasoain I, Fernández-Tomé P, et al. TACE/ADAM17-TNF-alpha pathway in rat cortical cultures after exposure to oxygen-glucose deprivation or glutamate. J Cereb Blood Flow Metab. 2002;22(5):576–585. doi:10.1097/00004647-200205000-00009

29. Bigdeli MR, Khoshbaten A. In vivo preconditioning with normobaric hyperoxia induces ischemic tolerance partly by triggering tumor necrosis factor-alpha converting enzyme/tumor necrosis factor-alpha/nuclear factor-kappaB. Neuroscience. 2008;153(3):671–678. doi:10.1016/j.neuroscience.2008.02.064

30. Chen C, Cui H, Li Z, Wang R, Zhou C. Normobaric oxygen for cerebral ischemic injury. Neural Regen Res. 2013;8(31):2885–2894. doi:10.3969/j.issn.1673-5374.2013.31.001

31. Schölzke MN, Schwaninger M. Transcriptional regulation of neurogenesis: potential mechanisms in cerebral ischemia. J Mol Med. 2007;85(6):577–588. doi:10.1007/s00109-007-0196-z

32. Zhang H, Vutskits L, Pepper MS, Kiss JZ. VEGF is a chemoattractant for FGF-2-stimulated neural progenitors. J Cell Biol. 2003;163(6):1375–1384. doi:10.1083/jcb.200308040

33. Bigdeli MR, Asheghabadi M, Khalili A. Time course of neuroprotection induced by normobaric hyperoxia in focal cerebral ischemia. Neurol Res. 2012;34(5):439–446. doi:10.1179/1743132812Y.0000000013

34. Esposito E, Mandeville ET, Hayakawa K, Singhal AB, Lo EH. Effects of normobaric oxygen on the progression of focal cerebral ischemia in rats. Exp Neurol. 2013;249:33–38. doi:10.1016/j.expneurol.2013.08.005

35. Flynn EP, Auer RN. Eubaric hyperoxemia and experimental cerebral infarction. Ann Neurol. 2002;52(5):566–572. doi:10.1002/ana.10322

36. Tiwari YV, Jiang Z, Sun Y, et al. Effects of stroke severity and treatment duration in normobaric hyperoxia treatment of ischemic stroke. Brain Res. 2016;1635:121–129. doi:10.1016/j.brainres.2016.01.030

37. Xu J, Zhang Y, Liang Z, et al. Normobaric hyperoxia retards the evolution of ischemic brain tissue toward infarction in a rat model of transient focal cerebral ischemia. Neurol Res. 2016;38(1):75–79. doi:10.1080/01616412.2015.1135558

38. Henninger N, Bouley J, Nelligan JM, Sicard KM, Fisher M. Normobaric hyperoxia delays perfusion/diffusion mismatch evolution, reduces infarct volume, and differentially affects neuronal cell death pathways after suture middle cerebral artery occlusion in rats. J Cereb Blood Flow Metab. 2007;27(9):1632–1642. doi:10.1038/sj.jcbfm.9600463

39. Kim HY, Singhal AB, Lo EH. Normobaric hyperoxia extends the reperfusion window in focal cerebral ischemia. Ann Neurol. 2005;57(4):571–575. doi:10.1002/ana.20430

40. Veltkamp R, Sun L, Herrmann O, et al. Oxygen therapy in permanent brain ischemia: potential and limitations. Brain Res. 2006;1107(1):185–191. doi:10.1016/j.brainres.2006.05.108

41. Singhal AB, Dijkhuizen RM, Rosen BR, Lo EH. Normobaric hyperoxia reduces MRI diffusion abnormalities and infarct size in experimental stroke. Neurology. 2002;58(6):945–952. doi:10.1212/WNL.58.6.945

42. Singhal AB, Benner T, Roccatagliata L, et al. A pilot study of normobaric oxygen therapy in acute ischemic stroke. Stroke. 2005;36(4):797–802. doi:10.1161/01.STR.0000158914.66827.2e

43. Singhal AB, Ratai E, Benner T, et al. Magnetic resonance spectroscopy study of oxygen therapy in ischemic stroke. Stroke. 2007;38(10):2851–2854. doi:10.1161/STROKEAHA.107.487280

44. Wu O, Lu J, Mandeville JB, et al. Dynamic functional cerebral blood volume responses to normobaric hyperoxia in acute ischemic stroke. J Cereb Blood Flow Metab. 2012;32(9):1800–1809. doi:10.1038/jcbfm.2012.87

45. Cheng Z, Li FW, Stone CR, et al. Normobaric oxygen therapy attenuates hyperglycolysis in ischemic stroke. Neural Regen Res. 2021;16(6):1017–1023. doi:10.4103/1673-5374.300452

46. Sun L, Strelow H, Mies G, Veltkamp R. Oxygen therapy improves energy metabolism in focal cerebral ischemia. Brain Res. 2011;1415:103–108. doi:10.1016/j.brainres.2011.07.064

47. Shin HK, Dunn AK, Jones PB, et al. Normobaric hyperoxia improves cerebral blood flow and oxygenation, and inhibits peri-infarct depolarizations in experimental focal ischaemia. Brain. 2007;130(Pt 6):1631–1642. doi:10.1093/brain/awm071

48. Shin HK, Oka F, Kim JH, Atochin D, Huang PL, Ayata C. Endothelial dysfunction abrogates the efficacy of normobaric hyperoxia in stroke. J Neurosci. 2014;34(46):15200–15207. doi:10.1523/JNEUROSCI.1110-14.2014

49. Walder CE, Green SP, Darbonne WC, et al. Ischemic stroke injury is reduced in mice lacking a functional NADPH oxidase. Stroke. 1997;28(11):2252–2258. doi:10.1161/01.STR.28.11.2252

50. Hirabayashi H, Takizawa S, Fukuyama N, Nakazawa H, Shinohara Y. Nitrotyrosine generation via inducible nitric oxide synthase in vascular wall in focal ischemia-reperfusion. Brain Res. 2000;852(2):319–325. doi:10.1016/S0006-8993(99)02117-4

51. Liu W, Chen Q, Liu J, Liu KJ. Normobaric hyperoxia protects the blood brain barrier through inhibiting Nox2 containing NADPH oxidase in ischemic stroke. Med Gas Res. 2011;1(1):22. doi:10.1186/2045-9912-1-22

52. Yuan Z, Pan R, Liu W, Liu KJ. Extended normobaric hyperoxia therapy yields greater neuroprotection for focal transient ischemia-reperfusion in rats. Med Gas Res. 2014;4:14. doi:10.1186/2045-9912-4-14

53. Liu W, Sood R, Chen Q, et al. Normobaric hyperoxia inhibits NADPH oxidase-mediated matrix metalloproteinase-9 induction in cerebral microvessels in experimental stroke. J Neurochem. 2008;107(5):1196–1205. doi:10.1111/j.1471-4159.2008.05664.x

54. Henninger N, Bratane BT, Bastan B, Bouley J, Fisher M. Normobaric hyperoxia and delayed tPA treatment in a rat embolic stroke model. J Cereb Blood Flow Metab. 2009;29(1):119–129. doi:10.1038/jcbfm.2008.104

55. Tang X, Liu KJ, Ramu J, Chen Q, Li T, Liu W. Inhibition of gp91(phox) contributes towards normobaric hyperoxia afforded neuroprotection in focal cerebral ischemia. Brain Res. 2010;1348:174–180. doi:10.1016/j.brainres.2010.05.082

56. Liu W, Hendren J, Qin XJ, Shen J, Liu KJ. Normobaric hyperoxia attenuates early blood-brain barrier disruption by inhibiting MMP-9-mediated occludin degradation in focal cerebral ischemia. J Neurochem. 2009;108(3):811–820. doi:10.1111/j.1471-4159.2008.05821.x

57. Singhal AB, Wang X, Sumii T, Mori T, Lo EH. Effects of normobaric hyperoxia in a rat model of focal cerebral ischemia-reperfusion. J Cereb Blood Flow Metab. 2002;22(7):861–868. doi:10.1097/00004647-200207000-00011

58. Shi S, Qi Z, Ma Q, et al. Normobaric hyperoxia reduces blood occludin fragments in rats and patients with acute ischemic stroke. Stroke. 2017;48(10):2848–2854. doi:10.1161/STROKEAHA.117.017713

59. Chern CM, Liou KT, Wang YH, Liao JF, Yen JC, Shen YC. Andrographolide inhibits PI3K/AKT-dependent NOX2 and iNOS expression protecting mice against hypoxia/ischemia-induced oxidative brain injury. Planta Med. 2011;77(15):1669–1679. doi:10.1055/s-0030-1271019

60. Woodfin A, Hu DE, Sarker M, Kurokawa T, Fraser P. Acute NADPH oxidase activation potentiates cerebrovascular permeability response to bradykinin in ischemia-reperfusion. Free Radic Biol Med. 2011;50(4):518–524. doi:10.1016/j.freeradbiomed.2010.12.010

61. Murotomi K, Takagi N, Mizutani R, et al. mGluR1 antagonist decreased NADPH oxidase activity and superoxide production after transient focal cerebral ischemia. J Neurochem. 2010;114(6):1711–1719. doi:10.1111/j.1471-4159.2010.06882.x

62. Liu C, Weaver J, Liu KJ. Rapid conditioning with oxygen oscillation: neuroprotection by intermittent normobaric hyperoxia after transient focal cerebral ischemia in rats. Stroke. 2012;43(1):220–226. doi:10.1161/STROKEAHA.111.625756

63. Yuan Z, Liu W, Liu B, Schnell A, Liu KJ. Normobaric hyperoxia delays and attenuates early nitric oxide production in focal cerebral ischemic rats. Brain Res. 2010;1352:248–254. doi:10.1016/j.brainres.2010.07.010

64. Qi Z, Yuan S, Liu KJ, Ji X. Normobaric hyperoxia plays a neuroprotective role after cerebral ischemia by maintaining the redox homeostasis and the level of connexin43 in astrocytes. CNS Neurosci Ther. 2022;28(10):1509–1518. doi:10.1111/cns.13875

65. Dong W, Qi Z, Liang J, et al. Reduction of zinc accumulation in mitochondria contributes to decreased cerebral ischemic injury by normobaric hyperoxia treatment in an experimental stroke model. Exp Neurol. 2015;272:181–189. doi:10.1016/j.expneurol.2015.04.005

66. Wang M, Geng X, Dandu C, Patel R, Ding Y. Normobaric Oxygen (NBO) therapy reduces cerebral ischemia/reperfusion injury through inhibition of early autophagy. Evid Based Complement Alternat Med. 2021;2021:7041290. doi:10.1155/2021/7041290

67. Begum G, Song S, Wang S, et al. Selective knockout of astrocytic Na(+) /H(+) exchanger isoform 1 reduces astrogliosis, BBB damage, infarction, and improves neurological function after ischemic stroke. Glia. 2018;66(1):126–144. doi:10.1002/glia.23232

68. Uria-Avellanal C, Robertson NJ. Na+/H+ exchangers and intracellular pH in perinatal brain injury. Transl Stroke Res. 2014;5(1):79–98. doi:10.1007/s12975-013-0322-x

69. Leng T, Shi Y, Xiong ZG, Sun D. Proton-sensitive cation channels and ion exchangers in ischemic brain injury: new therapeutic targets for stroke. Prog Neurobiol. 2014;115:189–209. doi:10.1016/j.pneurobio.2013.12.008

70. Assentoft M, Larsen BR, MacAulay N. Regulation and function of AQP4 in the central nervous system. Neurochem Res. 2015;40(12):2615–2627. doi:10.1007/s11064-015-1519-z

71. Tang G, Yang GY. Aquaporin-4: a potential therapeutic target for cerebral edema. Int J Mol Sci. 2016;17(10):1413. doi:10.3390/ijms17101413

72. Yang D, Ma L, Wang P, et al. Normobaric oxygen inhibits AQP4 and NHE1 expression in experimental focal ischemic stroke. Int J Mol Med. 2019;43(3):1193–1202. doi:10.3892/ijmm.2018.4037

73. Haelewyn B, Chazalviel L, Nicole O, Lecocq M, Risso JJ, Abraini JH. Moderately delayed post-insult treatment with normobaric hyperoxia reduces excitotoxin-induced neuronal degeneration but increases ischemia-induced brain damage. Med Gas Res. 2011;1(1):2. doi:10.1186/2045-9912-1-2

74. Li W, Qi Z, Ma Q, et al. Normobaric hyperoxia combined with endovascular treatment for patients with acute ischemic stroke: a randomized controlled clinical trial. Neurology. 2022;99(8):e824–e834. doi:10.1212/WNL.0000000000201592

75. Rønning OM, Guldvog B. Should stroke victims routinely receive supplemental oxygen? A quasi-randomized controlled trial. Stroke. 1999;30(10):2033–2037. doi:10.1161/01.STR.30.10.2033

76. Chiu EH, Liu CS, Tan TY, Chang KC. Venturi mask adjuvant oxygen therapy in severe acute ischemic stroke. Arch Neurol. 2006;63(5):741–744. doi:10.1001/archneur.63.5.741

77. Padma MV, Bhasin A, Bhatia R, et al. Normobaric oxygen therapy in acute ischemic stroke: a pilot study in Indian patients. Ann Indian Acad Neurol. 2010;13(4):284–288. doi:10.4103/0972-2327.74203

78. González RG, Hakimelahi R, Schaefer PW, Roccatagliata L, Sorensen AG, Singhal AB. Stability of large diffusion/perfusion mismatch in anterior circulation strokes for 4 or more hours. BMC Neurol. 2010;10:13. doi:10.1186/1471-2377-10-13

79. Roffe C, Ali K, Warusevitane A, et al. The SOS pilot study: a RCT of routine oxygen supplementation early after acute stroke--effect on recovery of neurological function at one week. PLoS One. 2011;6(5):e19113. doi:10.1371/journal.pone.0019113

80. Wu O, Benner T, Roccatagliata L, et al. Evaluating effects of normobaric oxygen therapy in acute stroke with MRI-based predictive models. Med Gas Res. 2012;2(1):5. doi:10.1186/2045-9912-2-5

81. Ali K, Warusevitane A, Lally F, et al. The stroke oxygen pilot study: a randomized controlled trial of the effects of routine oxygen supplementation early after acute stroke--effect on key outcomes at six months. PLoS One. 2014;8(6):e59274. doi:10.1371/journal.pone.0059274

82. Mazdeh M, Taher A, Torabian S, Seifirad S. Effects of normobaric hyperoxia in severe acute stroke: a randomized controlled clinical trial study. Acta Med Iran. 2015;53(11):676–680.

83. Li N, Wu L, Zhao W, et al. Efficacy and safety of normobaric hyperoxia combined with intravenous thrombolysis on acute ischemic stroke patients. Neurol Res. 2021;43(10):809–814. doi:10.1080/01616412.2021.1939234

84. Cheng Z, Geng X, Tong Y, et al. Adjuvant high-flow normobaric oxygen after mechanical thrombectomy for anterior circulation stroke: a randomized clinical trial. Neurotherapeutics. 2021;18(2):1188–1197. doi:10.1007/s13311-020-00979-3

85. Cheng Z, Gao J, Rajah GB, Geng X, Ding Y. Adjuvant high-flow normobaric oxygen after mechanical thrombectomy for posterior circulation stroke: a randomized clinical trial. J Neurol Sci. 2022;441:120350. doi:10.1016/j.jns.2022.120350

86. Ding J, Zhou D, Sui M, et al. The effect of normobaric oxygen in patients with acute stroke: a systematic review and meta-analysis. Neurol Res. 2018;40(6):433–444. doi:10.1080/01616412.2018.1454091

87. Mahmood A, Neilson S, Biswas V, Muir K. Normobaric oxygen therapy in acute stroke: a systematic review and meta-analysis. Cerebrovasc Dis. 2022;51(4):427–437. doi:10.1159/000521027

88. Li W, Lan J, Wei M, et al. Normobaric hyperoxia combined with endovascular treatment for acute ischaemic stroke in China (OPENS-2 trial): a multicentre, randomised, single-blind, sham-controlled trial. Lancet. 2025;405(10477):486–497. doi:10.1016/S0140-6736(24)02809-5

89. Liang J, Qi Z, Liu W, et al. Normobaric hyperoxia slows blood-brain barrier damage and expands the therapeutic time window for tissue-type plasminogen activator treatment in cerebral ischemia. Stroke. 2015;46(5):1344–1351. doi:10.1161/STROKEAHA.114.008599

90. Liu W, Hendren J, Qin XJ, Liu KJ. Normobaric hyperoxia reduces the neurovascular complications associated with delayed tissue plasminogen activator treatment in a rat model of focal cerebral ischemia. Stroke. 2009;40(7):2526–2531. doi:10.1161/STROKEAHA.108.545483

91. Ji Z, Liu K, Cai L, et al. Therapeutic effect of tPA in ischemic stroke is enhanced by its combination with normobaric oxygen and hypothermia or ethanol. Brain Res. 2015;1627:31–40. doi:10.1016/j.brainres.2015.08.019

92. Sun L, Zhou W, Mueller C, et al. Oxygen therapy reduces secondary hemorrhage after thrombolysis in thromboembolic cerebral ischemia. J Cereb Blood Flow Metab. 2010;30(9):1651–1660. doi:10.1038/jcbfm.2010.50

93. Fujiwara N, Murata Y, Arai K, et al. Combination therapy with normobaric oxygen (NBO) plus thrombolysis in experimental ischemic stroke. BMC Neurosci. 2009;10:79. doi:10.1186/1471-2202-10-79

94. David HN, Haelewyn B, Degoulet M, Colomb DG Jr, Risso JJ, Abraini JH. Prothrombolytic action of normobaric oxygen given alone or in combination with recombinant tissue-plasminogen activator in a rat model of thromboembolic stroke. J Appl Physiol. 2012;112(12):2068–2076. doi:10.1152/japplphysiol.00092.2012

95. Fisher M, Bastan B. Identifying and utilizing the ischemic penumbra. Neurology. 2012;79(13 Suppl 1):S79–85. doi:10.1212/WNL.0b013e3182695814

96. Li W, Hu W, Yuan S, et al. Enhancing blood-brain barrier integrity in patients with acute ischemic stroke via normobaric hyperoxia. J Am Heart Assoc. 2024;13(21):e036474. doi:10.1161/JAHA.124.036474

97. Li W, Wang S, Liu L, et al. Normobaric hyperoxia combined with endovascular treatment based on temporal gradient: a dose-escalation study. Stroke. 2024;55(6):1468–1476. doi:10.1161/STROKEAHA.123.046106

98. López HV, Vivas MF, Ruiz RN, et al. Association between post-procedural hyperoxia and poor functional outcome after mechanical thrombectomy for ischemic stroke: an observational study. Ann Intensive Care. 2019;9(1):59. doi:10.1186/s13613-019-0533-8

99. Parmar S, Moore-Langston S, Fredrickson V, et al. Neuroprotective mechanisms of oxygen and ethanol: a potential combination therapy in stroke. Curr Med Chem. 2015;22(10):1194–1204. doi:10.2174/0929867322666150114152945

100. Cai L, Thibodeau A, Peng C, et al. Combination therapy of normobaric oxygen with hypothermia or ethanol modulates pyruvate dehydrogenase complex in thromboembolic cerebral ischemia. J Neurosci Res. 2016;94(8):749–758. doi:10.1002/jnr.23740

101. Cai L, Stevenson J, Peng C, et al. Adjuvant therapies using normobaric oxygen with hypothermia or ethanol for reducing hyperglycolysis in thromboembolic cerebral ischemia. Neuroscience. 2016;318:45–57. doi:10.1016/j.neuroscience.2016.01.010

102. Cai L, Stevenson J, Geng X, et al. Combining normobaric oxygen with ethanol or hypothermia prevents brain damage from thromboembolic stroke via PKC-Akt-NOX Modulation. Mol Neurobiol. 2017;54(2):1263–1277. doi:10.1007/s12035-016-9695-7

103. Geng X, Sy CA, Kwiecien TD, et al. Reduced cerebral monocarboxylate transporters and lactate levels by ethanol and normobaric oxygen therapy in severe transient and permanent ischemic stroke. Brain Res. 2015;1603:65–75. doi:10.1016/j.brainres.2015.01.040

104. Cai L, Geng X, Hussain M, et al. Weight loss: indication of brain damage and effect of combined normobaric oxygen and ethanol therapy after stroke. Neurol Res. 2015;37(5):441–446. doi:10.1179/1743132815Y.0000000033

105. Geng X, Parmar S, Li X, et al. Reduced apoptosis by combining normobaric oxygenation with ethanol in transient ischemic stroke. Brain Res. 2013;1531:17–24. doi:10.1016/j.brainres.2013.07.051

106. Geng X, Elmadhoun O, Peng C, et al. Ethanol and normobaric oxygen: novel approach in modulating pyruvate dehydrogenase complex after severe transient and permanent ischemic stroke. Stroke. 2015;46(2):492–499. doi:10.1161/STROKEAHA.114.006994

107. Jin X, Liu J, Liu KJ, Rosenberg GA, Yang Y, Liu W. Normobaric hyperoxia combined with minocycline provides greater neuroprotection than either alone in transient focal cerebral ischemia. Exp Neurol. 2013;240:9–16. doi:10.1016/j.expneurol.2012.11.018

108. Liu Y, Liu WC, Sun Y, et al. Normobaric hyperoxia extends neuro- and vaso-protection of N-acetylcysteine in transient focal ischemia. Mol Neurobiol. 2017;54(5):3418–3427. doi:10.1007/s12035-016-9932-0

109. Nonaka Y, Koumura A, Hyakkoku K, et al. Combination treatment with normobaric hyperoxia and cilostazol protects mice against focal cerebral ischemia-induced neuronal damage better than each treatment alone. J Pharmacol Exp Ther. 2009;330(1):13–22. doi:10.1124/jpet.109.151548

110. Nonaka Y, Shimazawa M, Yoshimura S, Iwama T, Hara H. Combination effects of normobaric hyperoxia and edaravone on focal cerebral ischemia-induced neuronal damage in mice. Neurosci Lett. 2008;441(2):224–228. doi:10.1016/j.neulet.2008.06.033

111. Rodriguez P, Zhao J, Milman B, Tiwari YV, Duong TQ. Methylene blue and normobaric hyperoxia combination therapy in experimental ischemic stroke. Brain Behav. 2016;6(7):e00478. doi:10.1002/brb3.478

112. Beker MC, Caglayan AB, Kelestemur T, et al. Effects of normobaric oxygen and melatonin on reperfusion injury: role of cerebral microcirculation. Oncotarget. 2015;6(31):30604–30614. doi:10.18632/oncotarget.5773

113. Puy L, Parry-Jones AR, Sandset EC, Dowlatshahi D, Ziai W, Cordonnier C. Intracerebral haemorrhage. Nat Rev Dis Primers. 2023;9(1):14. doi:10.1038/s41572-023-00424-7

114. Zhou W, Marinescu M, Veltkamp R. Only very early oxygen therapy attenuates posthemorrhagic edema formation and blood-brain barrier disruption in murine intracerebral hemorrhage. Neurocrit Care. 2015;22(1):121–132. doi:10.1007/s12028-014-0013-9

115. Fujiwara N, Mandeville ET, Geng X, et al. Effect of normobaric oxygen therapy in a rat model of intracerebral hemorrhage. Stroke. 2011;42(5):1469–1472. doi:10.1161/STROKEAHA.110.593350

116. Yu Z, Chen LF, Tang L, Hu CL. Effects of recombinant adenovirus-mediated hypoxia-inducible factor-1alpha gene on proliferation and differentiation of endogenous neural stem cells in rats following intracerebral hemorrhage. Asian Pac J Trop Med. 2013;6(10):762–767. doi:10.1016/S1995-7645(13)60134-0

117. Zhao BQ, Wang S, Kim HY, et al. Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med. 2006;12(4):441–445. doi:10.1038/nm1387

118. You P, Lin M, Li K, Ye X, Zheng J. Normobaric oxygen therapy inhibits HIF-1α and VEGF expression in perihematoma and reduces neurological function defects. Neuroreport. 2016;27(5):329–336. doi:10.1097/WNR.0000000000000542

119. Zhong Q, Zhou Y, Ye W, Cai T, Zhang X, Deng DY. Hypoxia-inducible factor 1-α-AA-modified bone marrow stem cells protect PC12 cells from hypoxia-induced apoptosis, partially through VEGF/PI3K/Akt/FoxO1 pathway. Stem Cells Dev. 2012;21(14):2703–2717. doi:10.1089/scd.2011.0604

120. Wu M, Chen K, Zhao Y, et al. Normobaric hyperoxia alleviates complement C3-mediated synaptic pruning and brain injury after intracerebral hemorrhage. CNS Neurosci Ther. 2024;30(3):e14694. doi:10.1111/cns.14694

121. Zhou D, Meng R, Li SJ, et al. Advances in chronic cerebral circulation insufficiency. CNS Neurosci Ther. 2018;24(1):5–17. doi:10.1111/cns.12780

122. Wang Y, Zhao X, Liu L, et al. Prevalence and outcomes of symptomatic intracranial large artery stenoses and occlusions in China: the Chinese Intracranial Atherosclerosis (CICAS) Study. Stroke. 2014;45(3):663–669. doi:10.1161/STROKEAHA.113.003508

123. Safouris A, Hambye AS, Sculier C, et al. Chronic brain hypoperfusion due to multi-vessel extracranial atherosclerotic disease: a potentially reversible cause of cognitive impairment. J Alzheimers Dis. 2015;43(1):23–27. doi:10.3233/JAD-141203

124. Ding JY, Liu Y, Rajah GB, et al. Normobaric oxygen may correct chronic cerebral ischemia-mediated EEG anomalies. CNS Neurosci Ther. 2021;27(10):1214–1223. doi:10.1111/cns.13703

125. Bai C, Wang Z, Stone C, et al. Pathogenesis and management in cerebrovenous outflow disorders. Aging Dis. 2021;12(1):203–222. doi:10.14336/AD.2020.0404

126. Ding J, Guan J, Rajah G, et al. Clinical and neuroimaging correlates among cohorts of cerebral arteriostenosis, venostenosis and arterio-venous stenosis. Aging. 2019;11(23):11073–11083. doi:10.18632/aging.102511

127. Ding J, Guan J, Ji X, Meng R. Cerebral venous sinus stenosis may cause intracranial arterial hypoperfusion. Clin Neuroradiol. 2020;30(2):409–411. doi:10.1007/s00062-019-00833-w

128. Zhou D, Ding J, Asmaro K, et al. Clinical characteristics and neuroimaging findings in internal jugular venous outflow disturbance. Thromb Haemost. 2019;119(2):308–318. doi:10.1055/s-0038-1676815

129. Garaci FG, Marziali S, Meschini A, et al. Brain hemodynamic changes associated with chronic cerebrospinal venous insufficiency are not specific to multiple sclerosis and do not increase its severity. Radiology. 2012;265(1):233–239. doi:10.1148/radiol.12112245

130. Ding J, Liu Y, Li X, et al. Normobaric oxygen may ameliorate cerebral venous outflow disturbance-related neurological symptoms. Front Neurol. 2020;11:599985. doi:10.3389/fneur.2020.599985

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.