Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 1.
Published in final edited form as: Prog Neurobiol. 2014 Jan 7;0:246–269. doi: 10.1016/j.pneurobio.2013.12.007

Non-pharmaceutical therapies for stroke: Mechanisms and clinical implications

Fan Chen 1,#, Zhifeng Qi 1,#, Yuming Luo 1, Taylor Hinchliffe 2, Guanghong Ding 3, Ying Xia 2,*, Xunming Ji 1,*
PMCID: PMC3969942  NIHMSID: NIHMS562108  PMID: 24407111

Abstract

Stroke is deemed a worldwide leading cause of neurological disability and death, however, there is currently no promising pharmacotherapy for acute ischemic stroke aside from intravenous or intra-arterial thrombolysis. Yet because of the narrow therapeutic time window involved, thrombolytic application is very restricted in clinical settings. Accumulating data suggest that non-pharmaceutical therapies for stroke might provide new opportunities for stroke treatment. Here we review recent research progress in the mechanisms and clinical implications of non-pharmaceutical therapies, mainly including neuroprotective approaches such as hypothermia, ischemic/hypoxic conditioning, acupuncture, medical gases, transcranial laser therapy, etc. In addition, we briefly summarize mechanical endovascular recanalization devices and recovery devices for the treatment of the chronic phase of stroke and discuss the relative merits of these devices.

Keywords: non-pharmaceutical therapies, stroke, neuroprotection, acupuncture, hypothermia, brain conditioning, medical gases, transcranial laser therapy, mechanical endovascular recanalization

1. Introduction

Stroke, deemed a worldwide leading cause of death and neurological disability in both immature and adult subjects, continues to wreak physical, psychological and financial havoc in developing and developed nations alike. Instances of acute ischemic stroke result in heterogeneous changes in CBF (cerebral blood flow) and brain metabolism in the affected region. The human brain and its meshwork of roughly 100 billion interwoven neurons receives close to 15% of the body’s resting cardiac output while expending 20% of its oxygen, rendering the organ especially sensitive to such cases of hypoxic or ischemic insult (Clarke and Sokoloff, 1999; Fisher et al., 2009; Chao and Xia, 2010; Arai et al., 2011; Ding et al., 2012; Li et al., 2012a; Albert-Weißenbergeret al., 2013; He et al., 2013). As the brain is extremely sensitive to hypoxia or ischemia, protecting brain tissue from injury, simplified as “neuroprotection”, has therefore been a long sought after strategy in quelling physiological damage following stroke onset. Decades of research efforts have investigated over 1,000 pharmacological neuroprotectants, including excitatory amino acid antagonists, free radical scavengers, calcium channel blockers, and growth factors that are thought to target and correct different pathophysiological manifestations of ischemic brain injury. Unfortunately, almost all attempts at protecting the brain from ischemic injury have failed at making a successful transition into clinical use. The majority of these failures involved complex pharmaceutical targets and agents. Until now, intravenous rtPA (recombinant tissue plasminogen activator) is the only proven effective treatment in the acute setting. Most neuroprotectants alter only a single step in the broad cascade of biochemical events that lead to ischemic injury, and this over-specification may in part explain failure in the clinical setting (STAIR, 1999). Therefore, it is strongly suggested that stroke be approached with multiple, multifaceted neuroprotective methods capable of ameliorating a broader range of systems gone awry.

Non-pharmaceutical therapies are becoming more common and can be promptly initiated after stroke onset, marking them as ideal to combine with pharmaceutical or thrombolytic therapies which may further salvage affected brain tissue. Indeed, accumulating data suggest that “non-drug” approaches might provide new opportunities for stroke therapy, such as therapeutic hypothermia (Lakhan and Pamplona, 2012; Yenari and Han, 2012), ischemic/hypoxic conditioning (Liu et al., 2009b; Lim and Hausenloy, 2012), acupuncture (Guo et a.l, 2010; Xia et al., 2010; Xia et al., 2012), certain medical gases (Liu et al., 2011b; Sutherland et al., 2013) and other strategies.

In this review, we focus on the major clinical implications of non-pharmaceutical therapies for acute ischemic stroke and their underlying neuroprotective mechanisms. We also address the recent progress in mechanical endovascular recanalization and recovery devices for the chronic phase of stroke. Together, these non-pharmaceutical therapies present clinically translatable possibilities for both the acute and chronic stages of stroke recovery.

2. Hypothermia

The concept of cooling therapy comes from the phenomenon of hibernation in some mammalian species, which results in lowered body temperature and slowed metabolism. In addition to its use in maintaining organs at low temperatures to prolong transplantation viability, a hypothermic state - even a small, clinically feasible decline in body temperature - was found to prevent neuron death following global ischemia over 25 years ago (Busto et al., 1987). This finding has since prompted intense investigation into hypothermia as a neuroprotective approach, and has been shown in the laboratory to protect against experimental stroke through multiple mechanisms.

2.1. Mechanisms of hypothermia-induced protection against ischemic injury

Although the neuroprotective mechanisms of hypothermia have yet to be fully determined, several consequences of hypothermia seem highly relevant to decreasing the severity of ischemic brain damage, including slowed metabolism and the delay of pathophysiological progress, anti-inflammatory actions, a reduction of oxygen-based free radical production, decreased excitatory neurotransmitter release, prevention of blood-brain barrier disruption, gene expression alteration under ischemic conditions as well as the regulation of cell death and survival pathways.

2.1.1. Improvements in metabolism

Abnormal cellular metabolism, characterized primarily by the uncoupling of mitochondrial electron transport activity and rapid loss of ATP, is a major cause of brain injury under ischemic/hypoxic conditions (Sims and Muyderman, 2010). Thus, a logical neuroprotective approach would include either limiting or reversing such abnormal metabolic processes without causing irreversible cell death. In some models, hypothermia has been found to decrease metabolic rate and reduce blood flow in the brain (Erecinska et al., 2003). In such cases, low basal temperature was found to decrease brain oxygen consumption and glucose metabolism, and maintain valuable high-energy phosphate compounds such as ATP and pH in ischemic tissues, minimizing the downstream consequences of lactate overload and the development of acidosis (Schaller and Graf, 2003; Gotberg et al., 2010). Furthermore, in a gerbil model of global ischemia, normothermic recovery incurred a significant activity loss from non-synaptic mitochondrial electron transport enzyme complex I and synaptic and non-synaptic complexes II–III and IV (Canevari et al., 1999). Conversely, hypothermic (30°C) recovery led to respiration and mitochondrial enzymatic activity levels indistinguishable from sham-operated controls (Canevari et al., 1999). Although not all studies have indicated that hypothermia prevents ATP loss following ischemia, the recovery rate of ATP in hypothermic animals has been consistently observed in multiple models (Zhao et al., 2007a). The correlation between lowered temperature and the restoration of energetic capacity suggests that the beneficial effects of hypothermia involve, at least in part, mitochondrial function.

2.1.2. Inhibition of inflammatory mediators

Although inflammation is thought to partially contribute to tissue recovery and repair, inflammation can also exacerbate acute cerebral ischemic injury. The ischemic brain triggers innate immune responses that lead to the activation of microglia and circulating leukocytes, generating various molecules including ROS (reactive oxygen species), proteases and pro-inflammatory cytokines. Animal studies have shown that hypothermia may improve neurological outcome by inhibiting various pathological aspects of this immune response following brain ischemia and injury (Deng et al., 2003). Hypothermia suppressed neutrophil extravasation and microglia activation in the affected area and reduced the level of inflammatory mediators, including oxidative stress (Briyal and Gulati, 2010) and adhesion molecules (Choi et al., 2011). Additional studies found that levels of the proinflammatory cytokine IL-1β were notably reduced by local brain cooling (Fairchild et al., 2004; Wagner et al., 2006), which simultaneously decreased damage inflicted through vasogenic edema. These observations suggest the possibility that hypothermia may protect the blood brain barrier. However, it is important to note that anti-inflammatory cytokines such as IL-10 were also reduced by hypothermia, suggesting that hypothermia may have a more complex role in cytokine modulation (Matsui and Kakeda, 2008). Although much more investigation is required, suppressing inflammatory and downstream responses appears to serve as a neuroprotective mechanism in hypothermia therapy for cerebral ischemia.

2.1.3. Regulation of gene expression

Hypothermia has been found to affect gene expression under ischemic conditions. One study reported that hypothermia inactivated NF-κB (nuclear factorκB), a major transcription factor that regulates the expression of many inflammation-related genes. Accordingly, transcription levels of downstream transcriptional targets, including iNOS (inducible nitric oxide synthase) and TNF-α (tumor necrosis factor factor-α) were reduced by lowered temperatures (Han et al., 2003; Yenari and Han, 2006). In models of focal brain ischemia, hypothermia prevented nuclear NF-κB translocation and DNA binding by suppressing IκB phosphorylation and decreasing IKK activity (Han et al., 2003; Yenari and Han, 2006). As NF-κB also regulates genes involved in cell survival and growth, the comprehensive effect of hypothermia-induced suppression of NF-κB activity may be complex and is therefore still in the process of elucidation due to the different phases of ischemic injury; even the activation of NF-κB during these phases is believed to be a multistep process.

Overexpression of HSP70 (heat shock 70kDa protein) effectively reduced NF-κB activation in mice subjected to ischemic conditions (Zheng et al., 2008). In this study, HSP70 overexpression reduced the phosphorylation of IκB, thus prohibiting the entry of NF-κB into the nucleus and suppressed the expression of multiple genes controlled by NF-κB, including iNOS and TNF-α (Zheng et al., 2008). Endogenous HSP70 expression notably increased with hypothermia (Kaneko and Kibayashi, 2012), and thus could function as the mechanism leading to the suppression of NF-κB activation.

Excitotoxicity is an important theory that explains the pathophysiology of brain ischemia. The accumulation of excitotoxic amino acids (such as glutamate) is considered to be the base of excitotoxicity. Hypothermia limited calcium influx through AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid) channels, one of the major contributors to excitotoxicity under normothermic ischemic conditions. Under ischemic settings, GluR2 (glutamate receptor 2), a subunit of the AMPA receptor that limits calcium influx, is markedly down-regulated and leads to calcium overload, while GluR2 mRNA expression was only modestly reduced upon the addition of hypothermia. (Colbourne et al., 2003).

2.1.4. Balance between cell survival and death pathways

Hypothermia affects cell death processes in acute ischemic stroke models. Cell death pathways can be inhibited on several levels, including modulation of the expression of Bcl-2 (B-cell lymphoma-2) family members, the release of cytochrome c, the activation of caspases, or the activation of the FasL signaling pathway. In transient focal cerebral ischemia models, mild hypothermia significantly attenuated the release of cytochrome c in the brain, but did not alter the expression of Bcl-2 family proteins and caspase activation (Yenari et al., 2002). Hypothermia blocked ischemia-induced translocation of PKC (protein kinase C) δ to the mitochondria and nucleus (Shimohata et al., 2007); inhibition of PKCδ translocation to the mitochondria precludes its formation of reactive oxygen species and initiation of apoptosis. In models of both severe stroke, e.g., MCAO (middle cerebral artery occlusion) for 2 hours or longer, and global ischemia, hypothermia decreased the release of cytochrome c from the mitochondria (Zhao et al., 2007b; Yenari et al., 2002). Consistent with the improvement in stroke outcomes due to genetic or pharmacological disruption of the FAS/FASL pathway, hypothermia prevented FASL cleavage from the cell surface and decreased the amount of soluble FASL in cooled rodent brains (Liu et al., 2008a). The decreased level of soluble FASL was also associated with decreased caspase 8 activation, which occurs downstream of FAS activation (Liu et al., 2008a).

Hypothermia may also affect other molecules involved in apoptosis. The increased expression of HSP70 under hypothermic conditions may lead not only to the alteration of inflammatory gene expression as mentioned above, but may also serve as an important global mediator of cell death and survival pathways (Hu et al., 2013). PTEN (phosphatase and tensin homologue) is a tumor suppressor molecule with pro-apoptotic function when dephosphorylated. Under conditions of early hypothermia in which neuroprotection was observed, phosphorylated (inactive) PTEN levels were preserved, but not under delayed hypothermic conditions that failed to provide a neuroprotective effect (Lee et al., 2009). Although the relationship between phosphorylated PTEN levels and the suppression of disproportionate amounts of ROS is not well understood, the ROS scavenger S-PBN both reduced infarct volume by 57.2% and maintained levels of phosphorylated PTEN protein. It is possible, then, that hypothermia may act to suppress ROS, leading to preservation of PTEN phosphorylation.

2.1.5. Improved integrity of the blood–brain barrier

The BBB (blood brain barrier) is a physical and metabolic diffusion barrier between cerebral microvessels and the surrounding tissue and is essential for the maintenance of homeostasis and the normal function of the central nervous system (Liebner et al., 2011). Damage to the BBB is mediated by various factors, including free radical production, cytokines, and proteases (Rosenberg, 2012). In addition, structural components of the BBB such as fibronectin, laminin, collagen type IV and tight junction proteins are substrates for a class of extracellular proteinases called MMPs (matrix metalloproteinases) which are responsible for the breakdown of extracellular matrices. (Liu and Rosenberg, 2005). Focus on MMPs in models of cerebral ischemia has intensified due to the observations that activity and expression of MMP-2 and MMP-9 were significantly increased and closely related to BBB disruption, edema formation and intracranial hemorrhage (Yang and Rosenberg, 2011; Chaturvedi and Kaczmarek, 2013), suggesting that MMP activation may target BBB structural components and exacerbate acute brain injury.

Data generated from models of brain ischemia have shown that mild to moderate hypothermia protects the BBB and prevents edema formation (Preston and Webster, 2004; Lee et al., 2005; Kallmunzer et al., 2012). Lending support to the association of MMP activation and BBB damage in ischemic injury, hypothermia, which is protective against BBB damage and ischemia, was associated with a reduction in MMP proteolytic activity and degradation of various components of the extracellular matrix comprising the BBB, including vascular basement membrane proteins, agrin and laminin (Wagner et al., 2003).

Effects on the BBB extend well beyond the time frame of the hypothermic state. Even 5 days following the hypothermic regimen and focal cerebral ischemia, BBB integrity was preserved. On the cellular level, vascular morphology was improved by post-stroke hypothermic treatment, which led to less distortion of endothelial cells and maintained their association with the basement membrane (Duz et al., 2007). Hypothermic cooling also inhibited pericyte migration, which is increasingly recognized to have an important regulatory role in BBB integrity.

The above mentioned mechanisms for hypothermia-induced protection from ischemic brain injury are schematically shown in Figure 1.

Fig. 1. Potential mechanisms of hypothermia-induced protection from ischemic brain injury.

Fig. 1

Note that hypothermia therapy may reduce neurological injury through cellular and molecular regulation by decreasing metabolism and energy consumption, attenuating mitochondrial injury, inhibiting inflammatory mediators; decreasing MMPs proteolytic activity, inhibiting the FasL signaling pathway protecting the blood brain barrier, increasing HSP70 expression, inactivating NF-κB and downstream transcriptional targets, limiting calcium influx through AMPA, decreasing excitotoxicity, etc..

2.2. Hypothermia therapy for stroke in clinics

In cardiovascular medicine, the efficiency and safety of induced hypothermia in cardiac arrest has been clearly demonstrated (Zeiner et al., 2000; Bernard et al., 2002; Group, 2002). Indeed, mild hypothermia is recommended by the American Heart Association after ventricular fibrillation. However, the relationship between temperature and ischemic stroke has not yet been completely defined. Hyperthermia has been correlated with increased post-stroke morbidity and mortality in analyses of clinical studies (Hajat et al., 2000; Szczudlik et al., 2003). Conversely, preclinical animal models suggest that mild hypothermia is effective in neuroprotection in both focal transient occlusion models and global ischemia models (described above). Early hypothermia induction in patients with intracranial hematomas and severe traumatic brain injury was correlated with improved outcomes (Clifton et al., 2012), but the effects may have been due to better control of intracranial pressure rather than to hypothermia (Clifton et al., 2011). To date, no clear evidence supports the routine use of mild therapeutic hypothermia in acute stroke patients. Large randomized, controlled and multicenter clinical trials are still needed to ensure therapeutic effects and demonstrate the safety and feasibility of the therapy and standardize the therapeutic condition.

Despite the disappointing results from clinical trials, replicating the neuroprotection observed in experimental models of hypothermia may be dependent on optimal timing, target temperature, and duration of both hypothermia and occluded vessel reperfusion. Several preclinical studies have shown that hypothermia is effective when induced 1 hour after stroke (Zhang et al., 1993; Colbourne and Corbett, 1995) and that longer cooling increases its neuroprotective effects (Colbourne et al., 2000; Clark et al., 2009), but it is still debated as to how long after stroke hypothermia can be induced and remain neuroprotective. Moreover, the use of rapid cooling techniques may be combined with narrow time window thrombolysis/embolectomy which could further facilitate hypothermia-induced neuroprotection on stroke. However, even the rewarming process may be a sensitive variable to hypothermic neuroprotection, as it has been suggested that, in the presence of ongoing but potentially reversible intracranial hypertension, more prolonged hypothermia may be required and rewarming too quickly may result in temperature overshoot (Varon et al., 2012).

The methods to induce hypothermia can be classified into physical and pharmacological therapies, systemic and local therapies, and surface and endovascular therapies. All methods can be integrated with one another or combined with other neuroprotectants. Many of these methods possess significant caveats or confounds, which will be discussed below.

2.2.1 Surface cooling

Early surface cooling methods included convective air blankets, water mattresses, alcohol bathing, and ice packing. But maintenance of the target temperature using these methods is difficult to control, largely due to skin vasoconstriction and subsequent redistribution of blood flow. However, surface cooling has the considerable advantage of pre-hospital feasibility. The feasibility of surface cooling was addressed by the Copenhagen Stroke Study. In this study, 17 sober stroke patients tolerated mild hypothermia (mean of 35.5°C) for 6 hours with a forced air cooling blanket and were then gradually rewarmed over a period of 6 hours. Although the forced air method was well tolerated and free of confounds (e.g., anesthesia), no significant changes in short or long-term outcomes were found after accounting for factors such as age, gender, and stroke severity compared with 56 non-randomized control patients (Kammersgaard et al., 2000). Still, the well tolerated and anesthesia-free forced air method used, once further studied and refined, may eventually prove to be a highly efficient way of quelling stroke damage.

Localized surface cooling by the use of a cooling helmet is another method of inducing hypothermic conditions quickly and selectively without systemic complications (Wang et al., 2004). However, despite the convenience and portability, shaving is required to improve heat conduction. This delay may be critical when considering the limited therapeutic time for acute stroke patients. In addition, attaining deep brain temperatures are significantly less effective compared to lowering cortical temperatures. The distinction of ischemic injury typically centering in deep brain tissue thus limits the potential for therapeutic benefits of external surface cooling. However, penumbral cortical regions may still attain a therapeutic hypothermic temperature, and may improve functional, if not histological, outcomes.

Krieger et al conducted the COOL AID (Cooling for Acute Ischemic Brain Damage) pilot trial, of which was open design and nonrandomized. In total, 19 patients were eligible for the study, and 10 of them were treated with surface cooling (cooling blanket sandwiching) to induce moderate hypothermia (32±1°C) in combination with intravenous or intra-arterial thrombolysis. No statistical difference in mortality and complications between hypothermia patients and non-hypothermia patients occurred, suggesting that the procedure was well tolerated, but neurological outcomes were only slightly better in hypothermia patients. However, the lack of statistical significance could be attributed to several limiting factors, such as small sample size and open design (Krieger et al., 2001). Furthermore, the mean time from stroke onset to induction of hypothermia was 6.2 hours, and an additional 3.5 hours was required to achieve the target temperature. The extended timeframe to reach a hypothermic state following ischemia could also negatively impact any potential therapeutic benefit (Krieger et al., 2001).

2.2.2 Nasopharynx cooling

Nasopharynx hypothermia is another method that can be implemented as a prehospital hypothermic procedure. Circulating gases or fluids are either directly applied or contained within balloons by specific devices, and cooling is induced convectively or evaporatively. The cooled medium in the cavity induces direct cooling with close proximity to the brain by cooling the vascular supply in that region. This method cools more quickly than other surface cooling therapies, and is noninvasive and portable. However, similar to the helmet method, the cooling effect is localized, and nasopharyngeal cooling may preferentially cool the regions around the cribriform plate such as the basal frontal lobes, but other regions are only slightly affected at the same time (Mariak et al., 1999).

The nasopharynx hypothermia devices are various. For example, cold saline-filled thin-walled balloons (the QuickCool device, QuickCool AB, Lund, Sweden) or a specially designed nasal catheter that sprays highly evaporative substances (e.g., perfluorohexane (PFC), RhinoChill system (Benechill, Inc., San Diego, CA) can be inserted into the nasopharynx and lead to surface chilling. A randomized trial comparing the feasibility, safety and efficacy of the two methods (Cold Infusions vs. RhinoChill) for rapid cooling of stroke patients has been completed, but the result has not been published (ClinicalTrials.gov, Identifier: NCT01573117).

Another human trial of nasopharyngeal cooling is the Pre-ROSC (Return of Spontaneous Circulation) Intranasal Cooling Effectiveness study. In this clinical study, 83 of 182 patients were randomized to immediate pre-hospital nasopharyngeal cooling (Castren et al., 2010). The outcome data indicated that the nasopharyngeal cooling therapy improved neurological outcomes and decreased mortality. The most common side effect of treatment was reversible nasal discoloration (Castren et al., 2010).

2.2.3 Endovascular cooling

Endovascular cooling induces hypothermia by heat conduction mainly through cerebral blood flow, while local surface cooling does so mainly through brain tissue. Due to low conductivity of brain tissue, surface cooling requires an extended duration to reach the target temperature. Endovascular cooling therapy consists of intra-arterial infusion and intra-venous infusion. Because the cerebral blood flow is dependent upon the perfusion, recanalization therapies are essential. Intra-arterial infusion cooling can easily be combined with other endovascular interventions and has the advantage of cooling the highest risk tissue first. In spite of being capable of reaching the target temperature faster, endovascular cooling is invasive, non-removable and may cause significant complications (Ovesen et al., 2012).

The targeted and rapid localized cooling of high risk tissue can be attained by the intra-arterial infusion of cold fluids (such as recirculated blood and saline) directly into an ischemic arterial territory, using a microcatheter passed distal to an occlusion. In addition to directly targeting at risk tissue, the rapidity by which intra-arterial infusion cools the brain may shorten the onset and therapeutic duration of hypothermia as compared to the slower technique of surface cooling, thus avoiding potential complications that arise with an extended hypothermic condition. Furthermore, intra-arterial cooling could be easily integrated with other endovascular interventions

As opposed to the localized aspect of intra-arterial infusion, intra-venous infusion therapy induces hypothermia by infusion of cold solution through catheters placed into the inferior vena cava. The COOL AID II trial was a randomized pilot clinical feasibility trial of intra-venous endovascular cooling. Endovascular cooling and rewarming was induced by a device through the inferior vena cava. A core body temperature of 33 °C was targeted for 24 hours. 18 patients received endovascular cooling within 12 hours after a stroke, and 13 of them reached the target temperature of 33°C after an average of 77 minutes. As a control group, 22 patients received standard medical management. Clinical outcomes measured by the NIHSS (National Institutes of Health Stroke Scale) scores were similar in both groups. However, the infract volume measured by DWI (diffusion-weighted imaging) was lower in hypothermia patients (73%) than in non-hypothermia patients (124%) (De Georgia et al., 2004).

ICTuS-L (Intravenous Thrombolysis Plus Hypothermia for Acute Treatment of Ischemic Stroke) was a randomized, multicenter trial of endovascular cooling and intravenous tissue plasminogen activator in sober patients treated within 6 hours after ischemic stroke. The mean time to reach the target temperature (33°C) was 67 minutes and the duration was roughly 24 hours. Overall, 28 patients were randomized to receive hypothermia and 30 to receive normothermia. NIHSS clinical outcomes were found to be equivalent in both groups at 30 days, and at 3 months there was no statistically significant mRS (modified Rankin Scale) difference. The mortality and the rate of any intracerebral hemorrhage were also similar in both groups. Serious adverse events were more common in the hypothermia groups and the most common serious adverse event was pneumonia. This study demonstrated the feasibility and preliminary safety of combining endovascular hypothermia after stroke with intravenous thrombolysis, but did not confirm the hypothesis that cooling extends the time window for thrombolysis due to the small sample size used (Hemmen et al., 2010).

To circumvent complications (e.g., shivering, or measures to suppress shivering) associated with whole generalized hypothermia, recent research made use of selective ACP (antegrade cerebral perfusion) via the right axillary artery, allowing for hypothermic cooling of the brain only. During hypothermic circulatory arrest in 221 patients, moderate hypothermia induced by ACP during aortic arch surgery was associated with lower mortality and fewer neurologic sequelae than deep hypothermia compared to normothermic patients (Tsai et al., 2013).

2.2.4 Hypothermic combination therapy

Thus far, rtPA is the only proven therapy for ischemic stroke. The effect of hypothermia on thrombolysis is complicated, and may negatively affect each other. For example, recanalization brings increased blood flow and acceleration of heat condition, counteracting hypothermic cooling efforts. Likewise, the thrombolytic activity of rtPA decreased with cooler temperatures in an in vitro study (Yenari et al., 1995; Schwarzenberg et al., 1998). Still, there is no conclusive evidence suggesting a direct relationship between hypothermia and the incidence of hemorrhagic complications (Krieger et al., 2001; De Georgia et al., 2004). Several ongoing clinical trials are assessing the efficacy of therapeutic hypothermia in acute ischemic stroke in combination with other therapeutics. The ICTuS II/III study was designed as a prospective, randomized, single-blinded, multicenter phase II–III3 study in patients presenting within 3 h of stroke symptom onset who are also eligible for intravenous rtPA. Hypothermia is induced during thrombolysis by intravenous insertion of a catheter in the inferior vena cava and perfusion of iced saline to obtain a cooled state of 33 °C (Wu and Grotta, 2013). As an important note, the protocol calls for the use of anti-shivering interventions. Another ongoing trial assessing the possible synergy between hypothermia and thrombolysis is the EuroHYP-1, a European prospective open, randomized and controlled phase III clinical trial which induces hypothermia using external cooling pads in patients with acute ischemic stroke with or without receiving alteplase administration (Wu and Grotta, 2013). Dr. Grotta and colleagues also carried out several other clinical studies in their SPOTRIAS (Specialized Program of Translational Research in Acute Stroke) application. One of these included combining hypothermia with caffeinol, a neuroprotective combination developed in Dr. Grotta’s laboratory and tested in phase I and early phase II clinical trials. These evaluations demonstrated feasibility and safety.

2.2.5 Clinical issues

Despite the clinical feasibility of hypothermic induction, the procedure may lead to several complications or confounds that could affect both outcomes and interpretations. The primary medical complications of hypothermia are composed of cardiovascular complications, pulmonary complications, hematologic complications and immunologic complications. Unlike patients who are intubated and sedated following cardiac arrest or traumatic brain injury, acute ischemic stroke patients are awake, which in turn can impede rapid cooling because of discomfort and shivering. As a result, endotracheal intubation, sedation, pharmacological paralyzation, and anesthesia are induced to prevent shivering. Anti-shivering interventions may play an important role in improving patient tolerance, although pulmonary complications have been related to anti-shivering medications as well as to endotracheal intubation, and surface cooling methods alongside aggressive anti-shivering protocols may cause skin irritation and respiratory suppression (Doufas et al., 2003). Thus, precautionary measures should be implemented to minimize both discomfort and potential complications. Encouragingly, the effects of hypothermia were shown to not be affected by the administration of anti-shivering medication pethidine (Sena et al., 2012), but further studies are needed to develop better techniques to circumvent the use of additional measures used in minimizing complications.

In conclusion, although preclinical results are positive, the outcomes of translational research are still not stable. Appropriate parameters of cooling therapies for stroke still need to be verified. Larger and more definite clinical trials are critical for a better understanding of the full efficacy of hypothermia therapies. Furthermore, investigating the effects of hypothermia when combined with the wide variety of known and theoretical neuroprotectant and revascularization options must also continue. Future directions will push towards increasingly accurate brain-selective cooling and less invasive pharmacological cooling methods, altogether aimed at coming closer to a therapy that yields a balance between comfort, safety, and effective management of hypothermic conditions.

3. Ischemic/hypoxic conditioning

Training the brain to tolerate to ischemia, also called “conditioning”, is a profound neuroprotective strategy for stroke. Conditioning is a process by which previous exposure to a sublethal stress or stimulus confers resilience against a stronger stimulus. The concept of preconditioning was confirmed several decades ago by observing the protective effects of ischemia preconditioning on myocardial ischemia (Murry et al., 1986). The concept was further advanced to cerebral ischemia by the observation that brief bilateral carotid occlusion protected gerbil hippocampal CA1 pyramidal neurons against subsequent prolonged global ischemia (Kitagawa et al., 1991). In addition to these and others’ evidence that ischemic preconditioning protects against heart and brain ischemia, the phenomenon of ischemic preconditioning-induced protection extends now to the lung, kidney and liver as well (Fairbanks and Brambrink, 2010). Different methods that are able to develop conditioning have been emerging, including non-drug induced ischemic conditioning and hypoxic conditioning. Depending on the moment that conditioning is carried out, ischemic conditioning can be classified into preconditioning (before onset of reperfusion) and postconditioning (after ischemia and during reperfusion). However, it is not always feasible, practical, nor safe for stroke patients to receive conventional preconditioning or postconditioning treatment, which requires intra-arterial interventions of occluding and releasing the cerebral artery supplying the brain. RIPostC (Limb remote ischemic postconditioning) is a newly developed postconditioning procedure and is regarded to possess a high potential to become an efficacious protective strategy against brain ischemic injury (Lee et al., 2005). Here we summarize the common mechanism of ischemic/hypoxic conditioning.

3.1. Mechanism of ischemic/hypoxic conditioning

The mechanisms responsible for the induction and maintenance of ischemic/hypoxic tolerance in the brain are still undefined. However, laboratory studies suggest that the mechanisms for ischemic/hypoxic preconditioning involve a wide range of processes, including the improvement of cerebral blood flow, the up-regulation of protective membrane proteins, a reduction of inflammation, increased autophage function, and the regulation of survival/death signaling, among others.

3.1.1. Improvement in cerebral blood flow and vasculature

Acute ischemic stroke results in heterogeneous changes in CBF in the affected region. There is evidence showing that ischemic/hypoxic conditioning is neuroprotective due to improving CBF in ischemic stroke, likely through reduced reflow vasospasm (Gao et al., 2008a; Hasseldam et al., 2013). Using 14C iodoantipyrine autoradiography to measure CBF at varied occlusion intervals, a striking recovery of CBF during permanent MCAO in preconditioned spontaneously hypertensive rats was observed, presaging reductions in infarct volume (Zhao and Nowak, 2006). Specifically, global CBF detected in the contralateral cortex was better maintained following permanent occlusion in both preconditioned and sham animals, suggesting an effect of surgery (e.g., anesthesia) contributing to global CBF maintenance. In contrast, a progressive reperfusion of penumbra was detected in the ipsilateral cortex of the preconditioned group, independent of global perfusion changes (Zhao and Nowak, 2006).

The mechanisms of vascular alterations are still undetermined. Della-Morte et al have demonstrated that PKCε activated by ischemic preconditioning plays a pivotal role in neuroprotection by regulating CBF (Della-Morte et al., 2011). In addition, inhibition of adenosine transporter ENT1 (equilibrative nucleoside transporter 1) with propentofylline increased cerebral blood flow and re-established neuroprotection in hypoxic conditioning (Cui et al., 2013), indicating that maintenance of extracellular adenosine levels may be a mechanism by preconditioning to improve blood flow.

In addition to the observed alterations in blood flow, conditioning appears to directly preserve the vasculature at the molecular and cellular level. HPC (Hypoxic preconditioning) inhibited apoptotic cell death in human brain microvascular endothelial cells through PI3-kinase (phosphatidylinositol 3-kinase)/Akt and the inhibitor-of-apoptosis protein, survivin pathway (Zhang et al., 2007). We recently found that remote post-conditioning protected BBB integrity by diminishing MMP-9 expression and attenuating laminin and fibronectin degradation following experimental ischemic stroke (Ren et al., 2011; Liu et al., 2012c). SphK (Sphingosine kinase)-directed production of sphingosine-1-phosphate, which mediates peripheral vascular integrity via junctional protein regulation, was essential to hypoxic preconditioning-induced stroke protection. In wild-type mice, hypoxic preconditioning significantly strengthened the BBB integrity by increasing triton-insoluble claudin-5 and VE (vessel endothelium)-cadherin, while SphK2 knockout abolished HPC-induced BBB protection, with loss of VE-cadherin, occludin, and zona occludens-1 (Wacker et al., 2012).

3.1.2. Up-regulation of neuroprotective membrane functional proteins

Ischemic/hypoxic preconditioning can stimulate some membrane functional proteins to promote endogenous mechanisms of neuroprotection. Among them, the DOR (δ-opioid receptor) may play an important role (Ma et al., 2005; Zhang et al., 2006; Chao et al., 2008; Pang et al., 2009; Chao and Xia, 2010; He et al., 2012). We found that DOR signaling is oxygen-sensitive and up-regulated in both rapid (Zhang et al., 2006) and delayed (Ma et al., 2005) preconditioning. DOR signaling acts at multiple levels, including the regulation of survival and death signals (Ma et al., 2005), inhibition of glutamate toxicity (Zhang et al., 2006), and inhibition of Na+ influx through the membrane and reducing the increase in intracellular Ca2+, thus decreasing the excessive leakage of intracellular K+ in the cortex (Chao et al., 2007a). Such protection is dependent on a PKC-dependent and PKA (protein kinase A) -independent signaling pathway (Chao et al., 2008; Chao and Xia, 2010; He et al, 2013). In addition, other mechanisms of neuroprotection include the attenuation of neuronal transmission misbalance, increases in antioxidant capacity, regulation of specific gene and protein expression (Pang et al., 2009), and up-regulation of endogenous opioid release and/or DOR expression (He et al., 2012).

3.1.3. Attenuation of inflammation and the related events

Several examples of ischemic conditioning attenuate acute inflammatory processes. In the acute phase after stroke, inflammation can be detrimental to the neurological outcome. Ischemic postconditioning decreased myeloperoxidase activity and the expression of interleukin-1β and intercellular adhesion molecule 1 (Xing et al., 2008), and a long term protective paradigm of limb remote preconditioning inhibited activities of the proinflammatory galectin-9/Tim-3 pathway, iNOS, and nitrotyrosine (Wei et al., 2012). In situ superoxide detection using hydroethidine suggested that postconditioning attenuated superoxide products during early reperfusion after stroke (Zhao et al., 2006). In humans, remote limb conditioning reduced CD11b expression, as measured by flow cytometry and modified leukocyte inflammatory gene expression (Konstantinov et al., 2004). Ischemic conditioning also reduced neutrophil activation and inflammatory gene expression (Shimizu et al., 2010). The promotion of DOR by hypoxic/ischemic conditioning may also lead to suppression of inflammation, as we have recently found that activation of DOR attenuates ischemic increase in TNF-α (unpublished observation). Another mechanism of neuroprotection has been demonstrated by this pathway: a study indicated that ERK (extracellular regulated protein kinases) 1/2 activation induced by PKCε activation may result in NF-κB translocation to the nucleus followed by COX-2 (cyclooxygenase-2) expression, thus bringing about neuroprotection in mixed neuronal culture (Kim et al., 2010).

3.1.4. Regulation of cell survival/death signaling

Data derived from morphological, biochemical, and molecular genetic studies indicate that mitochondria constitute a convergence point for neurodegeneration. Mitochondrial dysfunction is also a prominent feature of cerebral ischemic stroke (Perez-Pinzon et al., 2012). Despite the fact that the precise molecular mechanisms underlying preconditioning-induced brain tolerance are still unclear, mitochondrial reactive oxygen species generation and mitochondrial ATP-sensitive potassium channel activation have been implicated in the preconditioning phenomenon. These pathways may be operative not only in brain but also in the brain endothelium. Ischemic/hypoxic preconditioning regulates the expression and translocation of Bcl-2 family proteins to mitochondria (Zhao et al., 2013), attenuating cytochrome c release and caspase-3 activation (Du et al., 2009). Excessive production of ROS and RNS (reactive nitrogen species) induced by ischemia is believed to perturb normal mitochondrial function and to initiate cellular death pathways. Despite the common association of ROS and toxicity, low levels of ROS production have been found to be protective and may serve as the trigger for activation of IPC-mediated neuroprotective pathways (Thompson et al., 2012). Furthermore, hypoxic preconditioning increased subcortical mitochondrial DNA content and mitochondrial density by a novel mechanism that requires nNOS (neuronal nitric oxide synthase), regulation of PGC-1α (α-subunit of peroxisome proliferators-activated receptor-γcoactivator-1) and CREB (cyclic adenosine monophosphate) response element binding protein (Gutsaeva et al., 2008). These data indicate that conditioning events can target mitochondria both in terms of modulation of cell death signaling as well as mitochondrial physiology.

Upstream of mitochondrial cell death signaling, multiple pathways are modulated by ischemic conditioning stimuli to counteract the detrimental signaling induced by severe brain injury. Key signal transduction pathways of IPC, such as activation of PKCε, MAPK (mitogen-activated protein kinase) and HIFs (hypoxia inducible factors) are likely involved in IPC-induced mitochondria mediated-neuroprotection (Della-Mortem et al., 2012). Recent findings suggest that signal transducers and activators of transcription, a family of transcription factors involved in many cellular activities, may be intimately involved in IPC-induced ischemic tolerance (Lin et al., 2011). In addition, sirtuins are stress-responsive enzymes which can be activated by IPC and may be linked to modulating protective pathways against oxidative stress, energy depletion, excitotoxicity, inflammation, DNA damage, and apoptosis. Although the functional roles of sirtuins under preconditioning scenarios are still being investigated, they possess unique roles in enhancing cellular function against stress-mediated damage, some of which have been associated with ischemia–reperfusion injury (Morris et al., 2011). The AKT/GSK3β (Glycogen synthase kinase-3β) pathway has been recognized as a protective pathway against cerebral ischemic injury. Remote limb conditioning activated and up-regulated the Akt pathway in cerebral ischemia models (Qi et al., 2012; Zhou et al., 2011a), and long term protection afforded by postconditioning was associated with Akt, MAPK (mitogen-activated protein kinase) and PKC (protein kinase C) signaling pathways (Gao et al., 2008b). Protection afforded by hypoxic preconditioning increased ERK and Bcl-2 activity, which counteracted the increased p38 MAPK activity and cytochrome c release observed following severe hypoxia (Ma et al., 2005). The cross-talk between ERK and p38 MAPKs displayed a “yin-yang” antagonism under the control of the DOR-G protein-PKC pathway (Ma et al., 2005). Taken together, the modulation of cell survival and death signaling by hypoxic/ischemic preconditioning appears to be capable of targeting multiple levels of signaling cascades.

3.1.5. Increased autophagy

The degradation of intracellular macromolecules or organelles (such as mitochondria) for subsequent reuse is a process that occurs under physiological conditions in order to maintain intracellular homeostasis. This process, known as autophagy, may also be involved in the protective properties of ischemic preconditioning and postconditioning (Balduini et al., 2012; Sheng et al., 2012a). In PC12 cells, ischemic preconditioning increased the generation and degradation of autophagosomes (Park et al., 2009). Inhibition of autophagy during either the lethal ischemic period or the reperfusion period following the lethal ischemic period significantly decreased the neuroprotective effects of ischemic preconditioning (Park et al., 2009). In a rat model of cerebral ischemic preconditioning, the autophagy inhibitors 3-Methyladenine and bafilomycin A1 suppressed neuroprotection induced by ischemic preconditioning, while rapamycin (an autophagy inducer) reduced infarct volume, brain edema and motor deficits induced by cerebral ischemia (Sheng et al., 2010). We recently reported the protective role of autophagy in rats exposed to limb ischemic postconditioning in the early phase of reperfusion. In this study, autophagy was promoted in an AKT-dependent manner, mainly in neurons in penumbral tissue (Qi et al., 2012). These observations indicate that autophagic activation during ischemic preconditioning may significantly contribute to tolerance toward a subsequent fatal ischemic insult.

3.1.6. Putative mechanisms underlying remote conditioning

The manner in which remote ischemic-reperfused tissue/organ can transmit protection to the brain or brain vasculature is still unclear. However, three theories have been formed based on the evidence from work in the myocardial and cerebral settings: i) humoral factors acting via the systemic circulation; ii) neurogenic transmission with involvement of muscle afferents and the autonomic nervous system; and iii) effects on leukocytes or circulating immune cells (Hess et al., 2013).

In summary, the mechanisms behind IPC/HPC protection from ischemic brain injury are schematically described in Figure 2.

Fig 2. Mechanisms of IPC/HPC protection from ischemic brain injury.

Fig 2

Note that IPC/HPC may block the biochemical cascades that cause brain injury following cerebral ischemia by improving CBF through PKC activation and adenosine transporter inhibition, inhibiting apoptosis through the PI3-kinase/Akt pathway, protecting BBB by diminishing MMP-9 and increasing SphK2, stimulating DOR signaling and thus regulating survival and death signals, inhibiting glutamate toxicity and maintaining ionic homeostasis, attenuating acute inflammatory processes though the PKC/ERK pathway, inhibiting mitochondrial cell death signaling through the AKT/PKC/MAPK/ERK pathway increasing autophagy.

3.2. Conditioning treatment for stroke in clinical trials

Although a substantial amount of preclinical data strongly indicates that ischemic postconditioning and preconditioning enhances brain and spinal cord tolerance to ischemia and reperfusion, the clinical data on their neuroprotective effects is scant (Koch et al., 2012; Narayanan et al., 2013).

IPC was first described in myocardial ischemia (Zhao et al., 2003), and to date, cardioprotection exerted by ischemic postconditioning has been reported in human trials (Hansen et al., 2010). The first clinical trial of ischemic postconditioning was performed following coronary angioplasty, where postconditioning was induced within 1 minute of reflow by 4 episodes of 1 minute inflation and 1 minute deflation of the angioplasty balloon (Staat et al., 2005). This study confirmed the concept in humans that coronary angioplasty postconditioning could protect the heart during acute myocardial infarction (Staat et al., 2005).

Our recent study correlated repetitive BAIPC (bilateral arm ischemic preconditioning) with reduced stroke recurrence in patients with IAS (intracranial arterial stenosis) (Meng et al., 2012). IAS patients (n=38) received BAIPC treatment along with standard medical treatment, while the control group (n=30) underwent standard medical treatment only. The incidence of recurrent stroke at 90 and 300 days in the BAIPC group was significantly lower compared to the control group, and the frequency of TIA (transient ischemic attack) onset in the duration of 16–30 days after the initiation of treatment was also statistically different. Interestingly, the average time to recovery (modified Rankin Scale score 0–1) was also shortened by BAIPC. Cerebral perfusion status, measured by SPECT and transcranial Doppler sonography, improved remarkably in BAIPC-treated brain than in that of controls (Meng et al., 2012). Further studies regarding the protective effects of remote limb ischemic preconditioning on acute cerebral infarction (ClinicalTrials.gov, Identifier: NCT01672515) are currently underway.

The endogenous protection conferred by conditioning protects the brain itself against future injury by adapting to small doses of harmful stimuli. After being described in myocardial ischemic preconditioning in a canine experimental model (Murry et al., 1986), its neuroprotective effect has also been repeatedly demonstrated in preclinical trials. The ability of the brain to mount an endogenous response to a conditioning stimuli that leads to subsequent protection against future injury has been described in many preclinical animal models, but may have also been indicated in clinical observations. Patients with previous, spontaneously occurring TIAs before cerebral infarction showed a more favorable outcome than those without TIAs. Although TIAs do not necessarily mimic the precise timing and intensity that has been deemed necessary in animal models of preconditioning, the study indicated that spontaneously occurring transient focal ischemia may induce a tolerance to subsequent permanent focal ischemia (Moncayo et al., 2000).

To date, the translation of preclinical ischemic conditioning protocols to a clinical application has been difficult in terms of both the methodology as well as the establishment of optimal timing. Most preclinical studies of remote ischemic conditioning use vessel clamps to induce hindlimb ischemia, while clinical trials to date have used tourniquets or blood pressure cuffs on the arm as a less invasive means to induce the preconditioning stimulus. In addition, the protection afforded by limb conditioning paradigms appears to be highly sensitive on the establishment of an optimal duration and repetitive ischemic cycles. Typically, preclinical studies have performed unilateral or bilateral femoral ischemia of varying time durations and cycle numbers. Factors such as the total duration of occlusion and the number of cycles need further optimization in clinical settings to determine both feasibility as well as targeted neuroprotective effects.

In designing ischemic postconditioning acute stroke clinical trials, the eligible population should be focused on patients with a high likelihood of reperfusion, either with IV rtPA or with mechanical intra-arterial interventions, since a primary effect of ischemic postconditioning is geared toward the minimization of reperfusion injury. Cardiac ischemia trials indicated that combining daily limb preconditioning with postconditioning may improve survival and left ventricular remodeling in the rat model (Wei et al., 2011), providing support for the application of both preconditioning plus additional postconditioning in the clinical setting. However, a recent clinical trial failed to show the superiority of the combination of limb preconditioning and postconditioning when compared to the control group (Hong et al., 2013). It is worth noting that other non-drug methods of inducing preconditioning, such as physical exercises in a broad sense, have been found to decrease ischemia/reperfusion brain injury in animal models (Li et al., 2004; Ding et al., 2006; Davis et al., 2007; Curry et al., 2010). Indeed, combination therapies with different conditioning may better salvage the ischemic tissue.

Large multicenter clinical studies are still needed to determine the impact of ischemic postconditioning and preconditioning on outcomes after stroke, though both postconditioning and preconditioning may be applied in a safe, economical, noninvasive and convenient fashion with definite neuroprotective potential. The difficulty in predicting the onset of ischemic stroke has limited the neuroprotective potential of ischemic preconditioning. Postconditioning therapy exerts a similar degree of effect compared to preconditioning (Zhao et al., 2006), and its potential feasibility is stronger for the application to both stroke treatment and prevention (Schaller et al., 2003). It is important to better define therapeutic windows by distinguishing different effects of preconditioning on ischemic cascades and CBF at various stages of stroke and the underlying mechanism in future studies. To increase the chance for success of future acute stroke clinical trials, further preclinical testing is needed according to STAIR (Stroke Therapy Academic Industry Roundtable) criteria.

4. Acupuncture

Acupuncture is one of the major modalities in traditional Chinese medicine (TCM). Throughout the long history of China, TCM has advocated for the use of acupuncture in ameliorating a variety of conditions including stroke and other neurological disorders (Xia et al., 2010; Xia et al., 2012). As recorded in the ancient literature, Huang Di Nei Jing (The Yellow Emperor’s Inner Classic, a product of various unknown authors in the Warring States Period, 475–221 BC), the clinical application of acupuncture on stroke can be traced back over 3000 years. Additional details were described in other historical TCM books such as Zhou Hou Fang (Handbook of Prescriptions for Emergencies) in the year 281 AD and Qian Jin Yao Fang (Essential Prescriptions Worth a Thousand Gold Pieces) in 680 AD (Chen et al., 2002; Guo et al., 2010; Zhou et al., 2011b). Since ancient times, the theory and practice of using acupuncture as a stroke treatment has been continually polished and refined throughout China and many other oriental countries. In fact, acupuncture is now recommended by the WHO (World Health Organization, 2003) as one of the alternative and complementary strategies against stroke.

Traditionally, acupuncture was delivered by the insertion of fine needles that were manually manipulated at different acupoints along the body. With the development of acupuncture theory and practice, many alternative techniques have been used to generate acupuncture signals, including acupoint stimulation with electrical currents, magnetic beads, or even by selective drugs. Due to its repeatability and feasibility of standardization, EA (electro-acupuncture) has become increasingly popular in both clinical and experimental studies (Xia et al., 2010; Xia et al., 2012).

The acupuncture modality is relatively convenient and cost-efficient with few side-effects. Acupuncture protocols have the potential to be optimized as an exceptionally useful therapeutic option for protecting the brain against stroke as an early intervention, especially when and where other treatments are not immediately available, such as at the home, during travel, or en route to a hospital. This additionally grants acupuncture therapies a chance of quelling stroke damage in a much more globalized context, extendable to regions with highly limited medical access.

4.1. Potential mechanisms for acupuncture protection against ischemic brain injury

In recent years, a variety of research with modern technologies has made major progress in elucidating the effects of acupuncture on stroke and the underlying mechanisms, especially in the acute ischemic condition. There is substantial evidence showing that either EA, which stimulates specific acupoints for an appropriate period of time with suitable current parameters, or manual acupuncture, can generate beneficial signals to the brain through peripheral nerve pathways (Guo et al., 2010; Xia et al., 2010; Xia et al., 2012). These signals lead to the regulation of cerebral blood flow and modulate the balance between survival and death signals at both cellular and molecular levels (Zhao et al., 2002; Tian et al., 2008; Zhou et al., 2008; Guo et al., 2010; Zhou et al., 2010; Zhou et al., 2011b; Xu et al., 2012; Li et al., 2012; Zhou et al., 2013a; Zhou et al., 2013b). In a side by side comparative study using the MCAO model, EA compared favorably to several pharmacological agents and Chinese herb medicines, such as taurine, “Salvia miltiorrhizae injection,“Qingkailing injection,” and “Xingnaojing injection” in terms of protective effects against ischemic injury (Zhao et al., 2008).

4.1.1. Increase of cerebral blood flow

The brain is highly susceptible to blood supply insufficiency, due in part to high energetic demands and to limited storage of energetic substrates and oxygen (Sung et al., 2008; Chao and Xia, 2010). The balance between energetic supply and demand can be disrupted by ischemic stress, leading to a disruption of ionic homeostasis and consequential dysfunction of neuronal excitability and synaptic transmission. The failure to maintain ionic homeostasis initiates multiple stress signaling pathways and the triggering of intracellular death signals. Consequently, prolonged ischemic periods lead to brain edema, infarction, neurological deficits, and even death. Therefore, restoration of blood flow to the infarct brain region has an invaluable impact on the treatment of stroke.

After optimizing EA at various acupoints with different current parameters on cerebral ischemia, we found that certain acupoints such as “Baihui” (Du 20) and “Shuigou” (Du 26) in the head and “Quchi (LI 11)” and “Neiguan” (PC 6) in the forelimb can significantly increase cerebral blood flow and reduce ischemic infarction, neurological deficit, and mortality following ischemic stroke. The effect on the blood flow is greatly dependent on the location stimulated, the stimulation intensity and frequency, and the duration of EA (Zhou et al., 2011b; Zhou et al., 2013a; Zhou et al., 2013b). Interestingly, the increase in blood flow occurred only in the ischemic, but not in the non-ischemic, brain (Zhou et al., 2011b).

The EA-induced increase in blood flow is isochronous to the current impulse and disappeared immediately after the termination of EA, suggesting a rapid-responsive mechanism (e.g., neural reflex) that contributes to the EA-induced increase in cerebral blood flow.

4.1.2. Regulation of neurotransmitters/modulators and membrane proteins

EA-induced increase in the blood flow likely exerts a major benefit to the ischemic brain. However, recent studies suggest parallel mechanisms contributing to EA-mediated brain protection following ischemic injury (Zhou et al., 2013b). There is substantial evidence demonstrating that acupuncture signals regulate many different neurotransmitters/modulators and membrane functional proteins (Wen, et al., 2010a; Wen, et al., 2010b; Liang and Xia, 2012; Xu, et al., 2012). Among them, DOR appears to play a unique role as an acupuncture-induced neuroprotective protein against cerebral ischemia.

As described in Section 3, DOR exerts neuroprotective effects in the brain (Chao and Xia, 2010; He et al., 2012). Activation of DOR protected against hypoxic and excitotoxic injury in cortical neurons (Zhang et al., 1999; Zhang et al., 2000; Zhang et al., 2002; Zhao et al., 2002; Ding et al., 2012). Pharmacological activation of DOR by the non-specific agonist DADLE ([D-Ala2, D-Leu5]-enkephalin) reduced glutamate-induced injury in neocortical neurons (Zhang et al., 2000), and was selectively blocked by δ-, but not by μ- or κ-, opioid receptor antagonists. Furthermore, we demonstrated that DOR provided neuroprotection against hypoxic/ischemic insults in various neural models, including neuronal cultures and brain slices subjected to hypoxia or oxygen-glucose deprivation, and in vivo brain exposed to cerebral ischemia (Ma et al., 2005; Zhang et al., 2006; Chao et al., 2007a; Chao et al., 2007b; Hong et al., 2007; Chao et al., 2008; Tian et al., 2008; Kang et al., 2009; Yang et al., 2009; Chao and Xia, 2010; He et al., 2012). Intracerebroventricular infusion of the DOR agonist TAN-67 significantly reduced the infarct volume and attenuated neurological deficits, while Naltrindole, a DOR antagonist, aggravated ischemic damage after forebrain ischemia in rats (Tian et al., 2008; Tian et al., 2013a). Similar data generated from other independent laboratories further demonstrated that DOR exerts neuroprotective effects against cerebral ischemic stress in animal models (Lim et al., 2004; Iwata et al., 2007; Su et al., 2007; Govindaswami et al., 2008; Borlongan et al., 2009). In addition, systemic administration of DOR agonist DADLE or Deltorphin-D (variant) reduced infarct volume after transient middle cerebral artery occlusion (MCAO) (Govindaswami et al., 2008; Borlongan et al., 2009).

DOR is highly distributed in the cortex and striatum, major regions targeted by stroke. Since abundant data in acupuncture research have shown that both manual acupuncture and EA can up-regulate endogenous opioid activity (Wen, et al., 2010b; Liang and Xia, 2012), it is very likely that acupuncture signal promotes DOR activity, thus protecting the brain from ischemic injury. In 2002, we showed that intracerebroventricular administration of Naltrindole, a DOR antagonist, largely reversed the EA-induced protection against the ischemic injury, which is the first evidence suggesting the role of DOR in the EA-induced protection against ischemic injury (Zhao, et al., 2002). Furthermore, we observed that EA rescued the level of DOR correlated with the reduction of ischemic infarction in the cortex (Tian et al., 2008a; 2008b). Intravenous injection of Naltrindole reversed the EA-induced protection (Zhou et al., 2013b). Dr. Xiong and his colleagues also demonstrated that DOR is involved in the EA-induced brain tolerance to cerebral ischemic injury (Xiong, et al., 2007; Li, et al., 2012). Intraperitoneal administration of Naltrindole abolished EA-induced protection against ischemic injury, while nor-BNI, a kappa-opioid receptor antagonist, did not significantly affect EA neuroprotection (Xiong, et al., 2007), demonstrating the specific role of DOR in brain protection against ischemic injury. Taken together, these various studies suggest that acupuncture may attenuate ischemic injury in the brain at least in part by the promotion of DOR signaling.

Other neurotransmitters/modulators and membrane functional proteins such as excitatory (e.g., glutamate) and inhibitory (e.g., GABA [gamma-aminobutyric acid]) amino acids, monoamines, neurotrophins such as BDNF (brain derived neurotrophic factor) and their receptors are also involved in acupuncture protection against ischemic brain injury, and have been recently reviewed (Guo, et al., 2010; Xu et al., 2012).

4.1.3. Promoting survival signal transduction

Though acupuncture may influence several neurotransmitters/modulators and accordingly regulate various survival/death signals, the DOR-mediated survival pathway may play a critical role in acupuncture-induced neuroprotection against ischemic brain injury. Recent studies have suggested that DOR activation can stabilize ionic homeostasis, suppress excitatory transmitter release, attenuate disrupted neuronal transmission, increase antioxidant capacity, inhibit apoptotic signals, promote pro-survival signaling and regulate specific gene and protein expression (Ma et al, 2005; Chao et al, 2007a; 2007b; 2008a; 2010a; 2012; He et al, 2013; Tian et al., 2013a; Tian et al., 2013b). For example, Bcl-2 and BDNF-TrkB survival signals are impaired/downregulated in hypoxic/ischemic conditions, while DOR activation rescues/upregulates them (Ma et al, 2005; Tian et al., 2013a; Tian et al., 2013b). Since EA promotes opioid activity in the brain (Wen, et al., 2010a; Wen, et al., 2010b; Liang and Xia, 2012; Xia et al, 2010; 2012) and rescued the level of DOR correlated with reduction of ischemic infarction in the cortex (Tian et al., 2008b), the EA-enhanced DOR activity very likely promotes Bcl-2 and BDNF-TrkB signaling, thereby leading to neuroprotection against ischemic injury.

In addition, the EA-mediated up-regulation of DOR signaling may regulate cellular and intracellular signal transduction at multiple levels, from membrane ionic homeostasis to gene expression and from neuronal integration to glial regulation, which have been largely summarized in recent reviews (Chao and Xia, 2010; He, et al., 2012). The EA-induced specific regulation of intracellular signal transduction has also been summarized recently (Guo, et al., 2010; Xu, et al., 2012). Therefore, the available data strongly suggest that acupuncture can promote survival signaling, while inhibiting death signals, thus rescuing the brain from ischemic injury.

Indeed, such DOR-mediated regulation is important for acupuncture to confer the neuroprotective effect because intracerebroventricular or intravenous injection of Naltrindole, a DOR agonist, largely reverses the EA-induced protection against ischemic brain injury (Zhao et al, 2002; Zhou et al, 2013b). On the other hand, an inappropriate EA stimulation that might not appropriately regulate the survival signaling in the brain does not induce any neuroprotective effect in spite that it still promotes an increase in cerebral blood flow (Zhou et al, 2013b).

4.2. Clinical practice

In past 10 years, many medical institutions, mostly in China and other oriental countries such as Korea, have reported the use of manual- or electro-acupuncture to treat stroke patients.

4.2.1. Controversy in clinical observations

Many of these reports demonstrated that acupuncture has a beneficial effect on patients with ischemic brain injury (e.g., stroke) in acute or chronic phases. After acupuncture therapy, patients showed improved recovery of motor function, reduced spasticity, relief of post-stroke depression, urinary incontinence or even reduction of infarction sizes (Hu et al., 1993; Johansson et al., 1993; Naeser et al., 1994; Si et al., 1998; Wong et al., 1999; Moon et al., 2003; Alexander et al., 2004; Schaechter et al., 2007; Bai et al., 2008; Liu et al., 2008b; Li et al., 2009b; Liu et al., 2009a; Chau et al., 2010).

However, despite these encouraging findings, the lack of strict random controls with appropriate statistical analysis of the data creates uncertainty of the reported efficacy in several of these clinical reports. Furthermore, several of the reports combined acupuncture with other medical treatments, which leads to difficulty in the correct evaluation of effects attributed to acupuncture rather than the adjunct therapy. Also, there is no standard/optimal protocol based on comparative studies in terms of therapeutic windows, acupoints, stimulation parameters, times and durations of treatment, etc. The treatments were mostly based on the personal experiences of the acupuncturists. Some clinical studies combined acupuncture and Chinese herbal medicine for the treatment of post-stroke motor dysfunction. However, most of the primary studies were inadequately designed trials characterized by unknown dropout rates and definitional vagueness in outcome measures. None of the studies approached important end points like death, survival times, rate of dependency, reduction in length of stay in hospital, etc. (Zhang et al., 2009). Combined, these limitations have led to the conclusion that data from the available trials are insufficient to guide continence care of adults after stroke (Thomas et al., 2008).

Although the difficulty of strictly controlling clinical research on patients with such serious disorders is understandable, these fatal issues greatly obstruct the promotion of clinical application for acupuncture in the treatment for stroke. Indeed, there are many reports, both dated and recent, that have indicated the uncertainty of acupuncture treatment for stroke and called for the necessity of more solid evidence in ascertaining the efficacy of acupuncture treatment for stroke (Gosman-Hedstrom et al., 1998; Liu et al., 2006; Hopwood et al., 2008; Kong et al., 2010). Thus, acupuncture, along with the other wide range of interventions pursued in clinical stroke research, require better quality evidence for successful clinical application (Thomas et al., 2008).

4.2.2. Potential solution of the controversy through research

The major controversy surrounding the efficacy of acupuncture in stroke treatment is likely attributed to the differences among various clinical studies in the selection of patients, in the windows of application, and/or in acupuncture methods including acupoints, stimulation parameters (or manner) of acupuncture stimulation and treatment times and durations. Indeed, the selection of proper stimulation approaches is a crucial factor that significantly influences the efficacy of acupuncture. We argue that improved preclinical research will help to establish the basic biology behind neuroprotection afforded by acupuncture as well as delineate therapeutic paradigms to improve standardization into clinical practice.

In the animal model, we observed that EA effectively reduced ischemic injury (Zhou et al., 2011b; Zhou et al., 2013a; Zhou et al., 2013b). When compared side by side to some protective drugs and compounds, EA showed the best effect in the reduction of ischemic infarction. However, we also found that optimized parameters for EA stimulation are extremely important. For example, the specific locations stimulated (acupoints) played a key role in the EA-induced increase in blood flow (Zhou et al., 2013a). Also, the stimulation parameters have a major impact on the EA-induced change in blood flow. No appreciable increase in the blood flow occurred using a stimulus with current intensity less than 0.8 mA, and the maximal effect was obtained by stimulating at 1 mA. However, the intensity at 1.2 mA failed to boost a further increase in the blood flow (Zhou et al., 2011b). In addition, the EA-induced increase in the blood flow was also dependent on stimulation frequency. EA given at a frequency of up to 40 Hz increased blood flow with a significant effect between 2 and 20 Hz. However, when the frequency was further increased to between 30 and 40 Hz, EA induced significantly less pronounced changes in blood flow. At frequencies at or above 40 Hz, only marginal or no increases in blood flow were observed (Zhou et al., 2011b). These results suggest that besides the locations stimulated (acupoints), the EA-induced increase in the blood flow is very sensitive to electrical frequency. Moreover, over-stimulation even at appropriate locations with optimal stimulation parameters may insult rather than protect the brain in spite that the stimulation still increases CBF (Zhou et al., 2013a). Based on our recent evidence, we hypothesized that non-optimal (e.g., over-stimulation) may trigger some injury factors in the brain, for instance, an anti-DOR mechanism. Therefore, an optimal condition is extremely important for ideal outcome in acupuncture treatment for stroke.

Interestingly, other investigators made similar observations in studies using different models. In a study of EA-induced hypoalgesia in healthy volunteers, subjects were randomized to receive different durations (0 min, 20 min, 30 min, or 40 min) of asynchronous EA stimulations and then subjected to a hypoalgesia test (experimental cold thermal pain threshold model). Thirty minutes of asynchronous EA stimulation yielded the greatest hypoalgesic effect compared with 0, 20, or 40min stimulations (Wang et al., 2009). Optimizing parameters for EA has also been noted to be critical in the cardiovascular system (Li and Longhurst, 2010).

Taken together, more preclinical translational research is needed to better define optimal conditions for maximal efficacy of acupuncture-induced protection from ischemic brain injury. Through it, we may generate valuable insight into applicable guidelines for an improved use of acupuncture for stroke treatment. Furthermore, such studies should establish a consensus on standardized relevant outcome measures in order to design and conduct appropriate randomized controlled trials that adopt those standards for evidence-based practices (Zhang et al., 2009; Asakawa and Xia, 2012).

5. Medical gases

Medical gases, another non-drug approach, are neuroprotective against acute ischemic stroke. These gases range from traditional gases (oxygen and nitrous oxide) to gases such as nitric oxide, carbon monoxide, and hydrogen sulfide (Liu et al., 2011b).

5.1. Oxygen

Hypoxia is a critical component contributing to neuronal death in stroke. Increasing brain tissue oxygenation has long been considered a logical strategy against cerebral ischemia. Oxygen, a commonly used therapeutic agent, has distinct advantages over pharmaceutical drugs (Liu et al., 2011b). The ease of diffusion across the BBB (Hawkins and Egleton, 2008) as well as the relatively safe, well tolerated, and minimal dose-limiting side-effects (Singhal et al., 2005b) position oxygen therapy to likely be effective at improving oxygen supply to brain tissue. Additionally, breakthroughs in chemical engineering have resulted in the creation of oxygen gas-filled microparticles that provide oxygen delivery through direct administration to the blood stream, leading to decreased levels of hypoxemia in rabbits (Kheir et al., 2012). The design of a protocol for testing any potential neuroprotective effects of these microparticles in a stroke model has yet to be developed.

5.1.1. NBO

Although HBO (hyperbaric oxygen therapy) has been extensively studied, NBO (normobaric hyperoxia therapy) has distinct advantages. As HBO requires a chamber or other equipment to create a hyperbaric atmosphere, NBO is simple to administer, inexpensive and can be started by paramedics after stroke onset en route to a hospital. NBO reduced ischemic brain injury and improved functional outcome in several animal models (Singhal et al., 2002a; Liu et al., 2006; Henninger et al., 2007; Yuan et al., 2010; Wu et al., 2012). The neuroprotective effects of NBO may be explained by many possible mechanisms, such as improved metabolism, reduced oxidative stress, and preservation of the BBB. These mechanisms are summarized in Figure 3.

Fig. 3. Possible pathways involved in oxygenation-induced brain protection in ischemia.

Fig. 3

Note that hyperoxia therapy may induce neuroprotection against acute ischemic stroke by improving tissue oxygenation and metabolism, reducing ischemia-induced acidosis and ATP depletion, up-expressing MMP-9, attenuating BBB leakage, inhibiting NADPH oxidase and decreasing free radical (ROS/RNS), thus reducing oxidative stress.

5.1.1.1. Improvement in tissue oxygenation and metabolism

The maintenance of tissue oxygenation is critical for neural cells and is severely affected by stroke. The blockage of blood flow during a cerebral ischemic event induced a rapid decrease of localized interstitial pO2 as measured by paramagnetic resonce oximetry (30% of pre-ischemic values in penumbra and 4% in the core during cerebral ischemia) (Liu et al., 2004), NBO treatment improved cerebral perfusion and oxygenation in ischemic regions as evidenced by 2-D multispectral reflectance imaging and laser speckle flowmetry, as well as inhibited peri-infarct depolarizations (Shin et al., 2007).

Abnormal metabolism has also been associated with brain injury following ischemic/hypoxic conditions, leading to tissue acidosis and ATP depletion. Ischemic-induced acidosis and ATP depletion in the border zones were attenuated by NBO in a model of focal cerebral ischemia (Sun et al., 2011). In stroke patients treated with inhalation oxygen at a flow rate of 45L/min through a face mask for 8 hours, lactate, a marker for aerobic metabolism, decreased during NBO administration (Singhal et al., 2007). The decrease in lactate suggests that aerobic metabolism can be improved following stroke by NBO. The minimization of energy metabolic dysfunction and the restoration of tissue oxygenation in the acute phase of stroke (i.e., en route to a hospital) might prolong the time window available for reperfusion therapies and the subsequent rescue of the penumbral region (Fisher and Bastan, 2012).

5.1.1.2. Reduction of oxidative stress

Oxidative stress is generated in tissues with abnormal oxygen levels and has been widely considered as one of the primary mechanisms leading to ischemic and/or reperfusion injury (Allen and Bayraktutan, 2009). Although one could hypothesize that oxygen therapy might increase the levels of reactive oxygen species, recent evidence indicates that a short duration of NBO treatment does not increase oxidative stress if started early after stroke onset. NBO treatment during focal cerebral ischemia-reperfusion not only did not increase oxidative stress, as measured by heme oxygenase-1 induction and protein carbonyl formation (Singhal et al., 2002b), but was associated with the reduction of superoxide production and 8-OHdG generation, a biomarker of oxidative DNA damage (Liu et al., 2006). These results indicate that, although NBO supplies oxygen to neural tissue, it appears to reduce oxidative stress. gp91phox is the catalytic subunit of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase, and a major source of reactive oxygen species. Pointing to a possible mechanism of how NBO may reduce oxidative stress, NBO inhibited the up-regulation of gp91phox (Liu et al., 2008c). In addition, genomic deletion of gp91phox led to a situation where NBO was no longer able to reduce BBB leakage following cerebral ischemia (Liu et al., 2011a). The effect of NBO on nitric oxide generation has also been examined following stroke. NBO administered during cerebral ischemia delayed and attenuated early nitric oxide generation, possibly through inhibiting nNOS (Yuan et al., 2010). In this study, ischemia caused a rapid production of NOx (nitrite plus nitrate), peaking at 10 min after stroke onset, and then gradually declining to the baseline level at 60 min. NBO delayed the NOx peak to 30 min and attenuated the total amount of NOx. Moreover, NBO showed a similar inhibitory effect on NOx and 3-nitrotyrosine production as that of the specific nNOS inhibitor 7-nitroindazole.

Together, these studies suggest that under appropriate conditions NBO treatment may not cause an observable increase in the production of ROS, but instead may actually decrease ROS generation when penumbral pO2 is maintained close to the pre-ischemic level.

5.1.1.3. Protection of the blood brain barrier

Induction of MMP-9 is highly associated with the post-ischemic breakdown of the blood brain barrier. NBO treatment inhibited MMP-9 induction in the ischemic brain, and was associated with reduced occludin degradation, Evan’s blue extravasation and hemispheric swelling (Liu et al., 2006; Liu et al., 2008c; Liu et al., 2009c), all of which suggesting that NBO treatment protects against BBB damage. The mechanism by which NBO suppresses MMP-9 and attenuates BBB damage appears to involve the above-mentioned inhibition of NADPH oxidase, because i) inhibition of NADPH oxidase with apocynin or knockout of gp91phox resulted in much smaller magnitudes in MMP-9 induction and BBB leakage in the ischemic brain (Liu et al., 2006; Liu et al., 2008c; Liu et al., 2011a), and ii) NBO did not further reduce the induction of MMP-9 when gp91phox was knocked out (Liu et al., 2011a). These results indicate that inhibition of NADPH oxidase-derived ROS production may be an important mechanism underlying NBO-afforded BBB protection. As the tightness of the BBB is crucial to ensure that the ischemic brain safely withstands the return of blood flow, early treatment with NBO may increase the therapeutic time window for thrombolysis and decrease the risk of hemorrhage by attenuating or slowing down the disruption of the BBB.

5.1.1.4. NBO plus rtPA thrombolysis

Both animal and human studies suggest that NBO slows down the process of cell death after stroke. The delay in cell death initiation by NBO may allow for an extension of the therapeutic time window for thrombolysis. The potential for combined NBO treatment with rtPA thrombolysis was recognized several years ago (Singhal et al., 2002a; Henninger and Fisher, 2006). Indeed, several preliminary safety studies have emerged, indicating that the combination of NBO with rtPA did not increase hemorrhage volume at 10 hours or occurrence of confluent petechial hemorrhages at 24 hours in a rat embolic stroke model (Henninger et al., 2009), nor did NBO increase cellular markers of superoxide generation or brain levels of MMP-9 (Kim et al., 2005). These data provide important initial evidence to support the feasibility of combination therapy with NBO and rtPA in ischemic stroke.

However, further studies have yielded inconsistent results as to the proposed synergistic relationship between NBO and rtPA. In one study, combined NBO and rtPA significantly reduced the mortality rate, brain edema, hemorrhage, and MMP-9 augmentation in a filament occlusion rat model with 5-hour ischemia followed by 19-hour reperfusion, as compared with rtPA alone (Liu et al., 2009b). In contrast, another study indicated that NBO itself achieved cerebral blood flow restoration equivalent to rtPA, possibly through direct interaction with endogenous tPA molecules, promoting endogenous fibrinolysis (David et al., 2012b). The combination of NBO and rtPA had no neuroprotective effect on ischemic brain damage in this study, and was concluded to possibly increase the risk of hemorrhage (David et al., 2012b). Further investigation is required to assess the effect of NBO and rtPA combinatorial strategies.

5.1.2. HBO

Although HBO lacks the ready accessibility to treatment compared to NBO, HBO may be even more potent than NBO at conferring ischemic protection (Liu et al., 2011b; Rockswold et al., 2010). Recent studies indicated that the maximal benefits from HBO therapy require early post-stroke administration, short durations, and tissue reperfusion (Veltkamp et al., 2005; Yang et al., 2010). From these studies and others, we have insight into critical issues surrounding HBO therapy, such as the therapeutic time window of HBO and the optimal chamber pressure, and from this, a multicenter HBO trial is currently being planned.

5.1.3. Gas-induced preconditioning

Intermittent hyperoxia has been shown to induce ischemic preconditioning. Compared with hypoxic preconditioning, hyperoxia preconditioning may be safer and more readily available in clinical practice. Consistent with a role for hyperoxia in establishing a preconditioned state, prior exposure to alveolar hyperoxia prevented the hypoxia-induced enhancement of bronchial reactivity to carbachol and histamine (D’Brot and Ahmed, 1991). These studies were carried forward into models of cerebral ischemia, where repeated exposure to HBO (100% oxygen at 2ATA for 1h each) increased the tolerance of the brain against ischemic neuronal damage, and induced HSP-72 expression (Wada et al., 1996). Since then, the protective effects of hyperoxia preconditioning, especially HBO preconditioning, have been investigated in many animal models. Studies have suggested that the protective effects of HBO preconditioning against ischemic injury involves the suppression of inflammatory responses, such as hemeoxygenase-1 (He et al., 2011; Liu et al., 2011c), HIF-1α(Peng et al., 2008), and COX-2 (cyclooxygenase-2) (Cheng et al., 2011). HBO preconditioning against ODG in cortical neuronal cultures activated the PPARγ (Peroxisome proliferator-activated receptor γ) pathway, which in turn increased downstream antioxidant enzymatic activities (Zeng et al., 2012). HBO preconditioning attenuated hemorrhagic transformation that correlated with decreased HIF-1α and MMP-2 and MMP-9 activity in hyperglycemic MCAO rats (Soejima et al., 2012; Soejima et al., 2013). HBO preconditioning also abated mitochondrial-mediated apoptosis in ischemic tissue (Li et al., 2009a), and was associated with autophagy activation (a vital cellular pathway for the degradation of intracellular abnormal mitochondria) following focal cerebral ischemia in rats (Yan et al., 2011).

Intermittent NBO has also been shown to induce ischemic conditioning. When compared to continuous NBO, intermittent NBO treatment (4 short cycles of intermittent NBO/air treatment) administered during ischemia provided similar neuroprotection when assessed at 24 hours after reperfusion, but greater neuroprotection when observed at 72 hours following reperfusion (Liu et al., 2012a). Thus, the extended timeframe to induce neuroprotection suggests that, besides providing oxygen to the ischemic tissue, intermittent NBO likely exerts its neuroprotection through other mechanisms, such as triggering a novel form of postconditioning, such as the attenuation of superoxide generation.

Bigdeli et al. investigated the neuroprotective effect of prolonged and intermittent hyperoxia preconditioning. Their results demonstrated that the neuroprotection of intermittent exposure was superior to that of prolonged exposure (24-hour exposure in this study was unacceptable) (Bigdeli et al., 2007).

5.1.4. Clinical trials using therapeutic oxygenation (Hyperoxia preconditioning)

Emerging data suggest that therapeutic oxygenation (normobaric and hyperbaric oxygen therapy) may have positive effects on the neuroprotection of acute ischemic stroke patients, and may even extend the narrow therapeutic time window for stroke thrombolysis as an adjunctive therapy with rtPA (Henninger and Fisher, 2006). To date, the neuroprotective effects of therapeutic oxygenation have been investigated in various models, including global ischemia, forebrain ischemia, surgical brain injury, ischemia-reperfusion injury, neonatal hypoxia-ischemia, intracerebral hemorrhage, and spinal cord ischemia.

It is important to note that supplemental oxygen is not equivalent to therapeutic oxygenation. Supplemental oxygen often requires inhaled oxygen concentrations approaching 100%, and, according to an early observational study, has been associated with a worse one year survival rate in patients with mild-to-moderate stroke (Ronning and Guldvog, 1999). Whether supplemental oxygen is beneficial to patients with severe strokes requires further research. However, supplemental oxygen is not recommended in non-hypoxic patients with acute ischemic stroke under current stroke guidelines (Jauch et al., 2013).

HBO has failed to show efficiency in three randomized clinical trials, and has been associated with severe limitations, including the possibilities of exacerbating oxygen free radical injury and incurring stroke secondary to air embolization (Anderson et al., 1991; Nighoghossian et al., 1995; Rusyniak et al., 2003). The relationship between HBO in acute ischemic stroke and outcomes of mortality, functional health, and adverse effects was explored by a meta-analysis of clinical studies from four randomized control trials. The study concluded that, based on the current evidence, a beneficial role of HBO for patients with stroke was not supported (Carson et al., 2005). As a result, HBO is not recommended for the treatment of acute stroke in the latest guidelines (Jauch et al., 2013). However, the strength of the conclusions from this meta-analysis was hampered by the determination that one controlled clinical trial and 17 observational studies were deemed to be of “poor quality” and limitations in sample size (Carson et al., 2005).

NBO has been shown to induce vasoconstriction and improve dynamic functional cerebral blood flow in salvageable tissue (Wu et al., 2012), as well as increasing O2 availability, reducing metabolic disturbances and protecting the blood brain barrier (Shin et al., 2007; Liu et al., 2011a). In addition, it is simple to administer, noninvasive, inexpensive, widely available, and can be started promptly after stroke onset. In 2005, a study at Massachusetts General Hospital investigated the effects of normobaric oxygen therapy in acute ischemic stroke; eight patients that received high-flow oxygen therapy (45 L/min for 8 h) were evaluated by stroke scale scores (NIHSS) and MRI scans (DWI lesion and the DWI-PWI mismatch). High-flow oxygen within 12 hours after onset of ischemic stroke was associated with a transient improvement of neurological impairment (Singhal et al., 2005a). With the addition of three more cases and assessments with multi-voxel magnetic resonance spectroscopic imaging and diffusion/perfusion MRI, NBO therapy decreased lactate levels and preserved N-acetylcysteine levels in the brain, suggesting that NBO improves aerobic metabolism and preserves neuronal integrity in the ischemic brain (Singhal et al., 2007). A subsequent clinical trial using MRI Voxel-based algorithms to evaluate NBO treatment in early phase showed that ischemic lesion growth is attenuated during NBO treatment at 4h (Liu et al., 2012b).

A larger scale trial was imitated in 2007 at Massachusetts General Hospital to compare the safety and therapeutic efficacy of NBO (started within 9 hours of symptom onset) to those of room air (30–45L/min for 8 h). After 85 enrollments, it was terminated because of the imbalance in deaths favoring control arm (ClinicalTrials.gov, Identifier: NCT00414726). In 2010, a randomized pilot study in India evaluated the effect of normobaric high-flow oxygen therapy in patients with acute ischemic stroke by the clinical (NIHSS, mRS) and radiological (MRI) measurements. Twenty patients were randomized to receive NBO. In contrast to the above studies, the result showed that NBO did not improve the clinical scores of stroke outcome in patients with acute ischemic stroke. Furthermore, adverse effects were observed, including cardiac/pulmonary side effects and free radical injury (Padma et al., 2010).

Taken together, the available preclinical and clinical data suggest that NBO may present a promising therapeutic strategy in acute ischemic stroke and TIA (Hadjiev and Mineva, 2009; Hadjiev and Mineva, 2010). Significant benefits may be achieved simply by administering pure oxygen via a face mask, instead of hyperbaric chambers. But its potential risk and curative effect need to be carefully evaluated. Further studies are needed to ensure the safety of NBO and investigate the optimal usage of NBO, including therapeutic time window, duration, flow rates, the combination of thrombolysis and the subtype of the ischemic stroke. Other associated therapies including the combination of minocycline, cilostazol and edaravone also require more clinical evidence (Nonaka et al., 2008; Nonaka et al., 2009; Jin et al., 2013). Lastly, prospective studies using modern neuroimaging methods are needed to assess the degree and durability of clinical benefits, especially the long-term benefits, from this treatment.

5.2. Other gases

In addition to oxygen, laboratory and clinical evidence has also found that other gases may play a beneficial role in the treatment of stroke. For example, the administration of xenon, argon and helium has resulted in effective neuroprotection in rat models of neonatal hypoxia/ischemia (Zhuang et al., 2012).

Emerging data support the hypothesis that hydrogen, which has the advantage of crossing the BBB easily, might act as a free radical scavenger of hydroxyl radicals, thus promoting neuroprotection. Hydrogen (2%) both reduced the amount of toxic hydroxyl radicals and significantly reduced infarct volume following transient MCAO in rats (Ohsawa et al., 2007). Studies using other rat models confirmed the effects of hydrogen on the improvement of neurological function (Cai et al., 2008; Chen et al., 2010; Matchett et al., 2009). A recent clinical study treated 13 cerebral ischemia patients with hydrogen inhalation and showed that 3–4% hydrogen inhalation over 30 mins was feasible and safe in stroke patients (Ono et al., 2012).

Helium was proposed as a therapeutic gas in 1934, and subsequently heliox (helium/oxygen gas mixture) was proposed as a potential therapy for upper and lower airway obstruction (Barach, 1934). In a side by side comparison of the efficacy of hyperoxia and that of heliox in a rat transient MCAO model, heliox was more effective than hyperoxia in reducing infarct volume and improving neurological deficits if initiated immediately after the onset of ischemia (Pan et al., 2011; Pan et al., 2007). The best protective outcome was observed when heliox (70% helium) was administered immediately after the occlusion (Pan et al., 2011; Pan et al., 2007). Additionally, the protective actions of heliox therapy have been demonstrated in traumatic brain injury, cardiac ischemia and hypoxia ischemia (Coburn et al., 2008; Pagel et al., 2007), but the exact mechanisms behind these protective effects are still unclear.

Xenon is used as an anesthetic gas, and because of its NMDA (N-methyl-D-aspartate) receptor inhibition properties, it has potential neuroprotective roles during instances of brain ischemia. The protective effects of xenon have been identified in various models of rodent hypoxic/ischemia (Natale et al., 2006; Dingley et al., 2008; Ma et al., 2009), but clinical evidence is still limited. In addition to its effects as a single therapeutic, xenon has also been noted to have synergistic/antagonistic neuroprotective effects when used with other therapeutic strategies. The combination of xenon and dexamethasone resulted in neuroprotection (Behrens et al., 1998), and a combination treatment of xenon and hypothermia in both rat and pig models was also neuroprotective (Fries et al., 2012; Sheng et al., 2012b). However, xenon can attenuate rtPA-mediated protection by acting as an inhibitor of rtPA (David et al., 2010), thus any clinical use of xenon would have to occur either in the absence or following rtPA administration.

The neuroprotective effects of argon have been supported by studies both in vitro and in vivo, and it carries the advantage of being both more affordable and common than xenon, while still capable of protecting against excitotoxic injury (Loetscher et al., 2009; Ryang et al., 2011; David et al., 2012a;). Argon is thought to induce GABA neurotransmitters and thus inhibit NMDA, but, as GABA receptor activation has additional downstream pathway influences, the protective mechanisms at work during argon administration have yet to be fully determined. Still, promising preclinical results for argon following ischemia suggest that it could be further examined as a potential neuroprotectant for ischemic stroke in a clinical setting.

Taken together, although the neuroprotective effects of these therapeutic gases have been proposed, the definite efficacy and mechanisms should be further validated through in-depth laboratory research and clinical trials.

6. Transcranial laser therapy

TLT (Transcranial laser therapy) has been investigated as a neuroprotective treatment for acute ischemic stroke over 10 years. It may promote functional and behavioral recovery via energy metabolism and by enhancing cerebral blood flow. Two clinical trials, NEST (NeuroThera Effectiveness and Safety Trial)-1 and NEST-2, have evaluated the use of TLT to promote clinical recovery in patients with acute ischemic stroke.

6.1. Mechanisms

Although the mechanism is still incompletely understood, specific wavelengths of near-infrared light can penetrate through the scalp, skull, and the surface of brain tissue, and then convert from light to bioenergy. First, the specific wavelengths (808nm) of light excite cuprous moieties of the mitochondrial cytochrome C oxidase enzyme. This energy absorption within mitochondria increases ATP, leading to significant improvements in energy metabolism and cell viability. Such maintenance of homeostatic cellular functions helps to prevent the spread of ischemia-induced death within the penumbra (Mochizuki-Oda et al., 2002; Desmet et al., 2006; Lapchak and De Taboada, 2010). The biological effects of TLT were shown to be specific to the wavelength and not due to thermal effects of the laser beam (Lapchak, 2010). Other mechanisms may contribute to TLT-induced neuroprotection, such as the promotion of neurogenesis, the production of endogenous neurotrophic factors and the increase of CBF alongside the concentration of cortical NO (Leung et al., 2002; Oron et al., 2006; Uozumi et al., 2010). Taken together, TLT imposes neuroprotective effects by inducing photobiological stimulation to increase mitochondrial energy supply and CBF.

6.2. Clinical trials

Two randomized double-blind clinical trials with TLT induced by the NeuroThera device (Photothera, Carlsbad, CA) using near-infrared light with a wavelength of 808nm have been completed with reproducible and measurable clinical improvement in acute ischemic stroke patients (Lampl et al., 2007; Zivin et al., 2009), A third trial is currently being developed (Zivin et al., 2012).

The NEST-1 was a prospective, double-blinded, randomized, placebo-controlled, multicenter trial involving 120 subjects with strokes who were ineligible for intravenous thrombolysis. In 120 patients, 79 patients received TLT therapy that used low-energy (10mW/cm2) lasers at 20 predetermined locations on the scalp with 2 min of irradiation per site, over 20 sites, within 24 hours (mean application at 16 hours) after stroke onset. Significantly better neural function outcome (mRS and NIHSS) in the TLT-treated group were observed compared to the control group. In addition, there was no difference in mortality. All evidence indicated the safety and effectiveness of the treatment for ischemic stroke in humans (Lampl et al., 2007).

A larger follow-up phase III trial, NEST-2, used an identical procedure as described for NEST-1. In 660 patients, 331 patients who were accorded with the specific criterion received TLT within an average of 14.6 h post-stroke, and failed to find the same significant effectiveness in NEST-1. However, it confirmed the safety of the therapy and the suitable range of stroke patients (Zivin et al., 2009).

The NEST-3 trial (ClinicalTrials.gov, Identifier: NCT01120301), with the purpose of demonstrating the safety and efficacy of TLT in the treatment of acute ischemic stroke by assessing the mRS at 90 days post stroke, is a double-blind, randomized, sham-controlled, parallel group, multicenter, pivotal study and planned to enroll 1000 subjects. (Zivin et al., 2012). But NEST-3 failed to reach significance upon interim analysis in October 2012. The independent Data Monitoring Committee (DMC) found that the trial failed to pass a pre-specified hurdle for efficacy in a futility analysis performed on greater than 550 randomized patients who had completed their Day 90 evaluation. Upon unblinded review of the data, the DMC identified no safety concerns with the therapy. But no efficacy was detected either. So enrollment into the NEST-3 study was suspended and PhotoThera (the sponsoring company) has closed and dissolved.

Lapchak et al used a rabbit large clot embolic stroke model to determine the safety TLT in combination with rtPA, and demonstrated that TLT did not significantly affect hemorrhage rate, hemorrhage volume and the 24-hour survival rate in embolized rabbits (Lapchak et al., 2008). Future clinical trials with an enrollment of patients receiving rtPA should be applied to confirm the neuroprotection of TLT.

Since the efficiency of TLT therapy is still in question, further studies are likely to shed some light on the benefits and shortcomings of TLT therapy. The thermal effects of the laser beam may also affect safety and efficiency. Furthermore, electromagnetic wave energy is mainly concentrated on cortical tissue and cannot reach the deep brain parenchyma. Thus, the application of TLT in acute ischemic stroke is still in its early stages, and larger and more specific clinical trials are required to confirm optimal laser settings, patient population, therapeutic time windows, as well as to develop more advanced devices, all aimed towards creating a new paradigm for the treatment of acute ischemic stroke.

7. Mechanical Endovascular Recanalization

Mechanical endovascular recanalization therapies rehabilitate cerebral blood flow in occluded arteries, including thrombectomy devices which extract, disrupt and aspirate clots, balloon angioplasty or stents which displace clots to the periphery of vessels, and augmenting perfusion devices. These therapies require tailor made devices, qualified organizations, competent medical staff, and above all, timely treatment.

The common devices of recanalization include the Merci Retriever, Penumbra System, Phenox Retriever, Catch Device, Solitaire Device, and the Trevo Device. As indicated by the FDA (Food and Drug Administration), Merci is used for restore neurovascular flow, Penumbra is used for revascularization for acute stroke, and Solitaire and Trevo are approved for intracranial clot removal.

Two small scale trials indicated that the Solitaire Device and Trevo Device show better safety and efficacy than the Merci Retrieval System in endovascular recanalization in acute ischemic stroke (Nogueira et al., 2012; Saver et al., 2012). The latest guidelines recommend that, although the usefulness of the Merci, Penumbra System, Solitaire FR, and Trevo thrombectomy devices is certain, proven efficacy still needs more clinical evidence to improve patient outcomes. The usefulness of other mechanical thrombectomy devices as well as emergent intracranial angioplasty/stenting have not been established (Jauch et al., 2013).

Research on ultrasound enhanced recanalization is also ongoing. Owing to acoustic cavitation, ultrasound devices may enhance thrombolysis and improve recanalization rates compared to IV rtPA alone, thereby having the potential to improve the safety and efficacy of rtPA treatment. Adjuvant ultrasound using microbubbles enhanced rtPA thrombolysis in vitro and in vivo (Hitchcock and Holland, 2010; Nolte et al., 2013). By using a combination of ultrasound and endovascular techniques, the ultrasound transducer can be placed at the tip of the endovascular catheter can thus circumvent interference by bone (Tachibana et al., 1999). A SPOTRIAS study carried out by Dr. Grotta’s group is the development of a hands-free transcranial ultrasound unit, and its safety and efficacy is currently being tested in a phase 3 international prospective randomized study (NIH RePORT Project Number: 5P50NS044227-10). If positive, ultrasound enhanced thrombolysis could become widely applicable wherever IV rtPA is given.

Recently, several studies regarding the use of endovascular treatment for acute ischemic stroke with large artery occlusion yielded negative results, and have presented additional challenges to the creation of effective stroke therapies. The IMS III (interventional management of Stroke III) was a phase III, randomized, multicenter, open label, 656 subject (434 patients to endovascular therapy and 222 to intravenous rtPA alone) clinical trial (2006–2012). This study randomly assigned eligible patients who had received intravenous rtPA within 3 hours after symptom onset to receive additional endovascular therapy (Merci, Penumbra system, Solitarie FR) or intravenous rtPA alone, and compared the mRS and NIHSS scores between the combined therapy group and the intravenous rtPA alone group. Although the trial demonstrated similar safety outcomes, no significant difference in functional independence with endovascular therapy after intravenous rtPA was found, as compared with intravenous rtPA alone (Broderick et al., 2013).

The SYNTHESIS Expansion was a multicenter, open-treatment clinical trial with a blinded end point. 362 patients (181 patients received endovascular therapy and 181 intravenous rtPA) with acute ischemic stroke within 4.5 hours after stroke onset were randomly assigned to study groups (2008–2012). In this study, endovascular therapy did not appear superior to standard treatment with intravenous rtPA in safety and efficacy (Ciccone et al., 2013).

The MR RESCUE (mechanical retrieval and recanalization of stroke clots using embolectomy) was a phase IIb, randomized, controlled, open-label (blinded outcome), and multicenter trial (2004–2011). Randomly assigned patients within eight hours after the onset of large-vessel, anterior-circulation strokes were subjected to mechanical embolectomy (Merci Retriever or Penumbra System) or received standard care. All patients were stratified according to the existence of penumbra as measured by pretreatment computed tomography or magnetic resonance imaging of the brain. The presence of a favorable penumbral pattern on neuroimaging did not lead to a differentiation of patients who might benefit more from endovascular therapy for acute ischemic stroke. (Kidwell et al., 2013). In addition, embolectomy was not found to improve outcomes over the standard care.

Although the conclusions above are consistent and argue against the earlier hypothesis that endovascular therapy may be associated with improved outcomes, there are several limitations of these studies that may affect the strength of the conclusions. First, efficacy is strongly related to the rapidity of initiation of treatment, and the time of interventional therapies in these studies was delayed as compared with rtPA therapy. The minimization of any delays to endovascular therapy initiation could significantly affect the results. Secondly, the time spans of these studies are long, and there were advances in techniques and clinical practices during the study period. Therefore, the efficacy of the new devices and as compared with intravenous rtPA alone remains insufficient. Third, the neuroimaging came from a single time point and may have changed by the time of recanalization in patients undergoing embolectomy. Follow-up imaging was also not available for all patients. The latest imaging technologies and real-time analysis will be helpful to provide more accurate and credible results. In addition, the lack of a significant benefit of endovascular therapy may be related to the counteraction which is brought about by the operation, and unnecessary damage may be minimized by the accumulation of operational experience. However, facing the currently high rate of recanalization with endovascular treatment, overly accelerated steps should be halted in order to comprehensively theorize and deliberate over which future directions to take with endovascular mechanical recanalization therapy.

8. Recovery devices for chronic phase of stroke

Technology such as robotic devices, repetitive transcranial magnetic stimulation, functional electrical stimulation, vagus nerve stimulation, and transcranial direct current stimulation aims to improve recovery in the chronic phase of stroke. These devices are designed and developed to improve speech, motor and sensory function by facilitating brain function recovery and to preventing disuse myophagism. We divided them into two categories, i.e., the stimulus supporting devices and the exercise recovery devices.

The stimulus supporting devices contain FES (Functional Electrical Stimulation), rTMS (repetitive Transcranial Magnetic Stimulation), VNS (Vagus Nerve Stimulation), and tDCS (transcranial Direct Current Stimulation) (Wilkie et al., 2012; Kondo et al., 2012; Bastani and Jaberzadeh, 2012; Khodaparast et al., 2013). The FES devices facilitate motor ability and decrease activity limitation caused by stroke. The rTMS is a noninvasive method of restoring excitability and voluntary control of down-regulated neurons in the stroke hemisphere. It has been shown to be effective in the setting of low-frequency stimulation. VNS significantly reduces the extent of stroke-induced lesion of brain parenchyma and results in movement during rehabilitation. The tDCS is used to polarize underlying brain regions through the application of weak direct currents by electrodes on the scalp. The current regulates the neuronal excitability to facilitate cortical function.

Exercise recovery devices are deemed as bioengineered devices that focus on extremity impairment reduction after training. Meanwhile, they can measure motor performance objectively and will contribute to a detailed phenotype of stroke recovery. For instance, the AMES (assisted movement with enhanced sensation) device is designed to produce functional cortical changes by assisting the subject as the patient attempts to move the limb (assisted movement) and enhancing movement sensation by vibrating the muscles during movement (enhanced sensation). The training performs more effectively when in combination with EMG Biofeedback (Cordo et al., 2009).

The BCI (Brain Computer Interface) device for stroke rehabilitation has been developed to assist people with motor disability. It works by detecting the motor intent of the user from electroencephalogram signals to drive the robotic rehabilitation via Haptic Knob. The developmental direction of this device is moving towards decreasing electrode interference and improving the accuracy of results (Daly et al., 2009; Tam et al., 2011).

9. Concluding remarks

Stroke is a serious neurological condition involved in complex pathophysiological mechanisms at multiple levels (Clarke and Sokoloff, 1999; Chao and Xia, 2010; Puyal et al, 2013; He et al, 2013). A single pharmaceutical drug may not work properly at all critical levels in ischemic brain injury. This may be one of the key reasons behind the past unsuccessful efforts to achieve therapeutic goals through a pharmacological means. In contrast, non-pharmaceutical treatment such as hypothermia, preconditioning and acupuncture may induce a comprehensive effect on the ischemic brain at different levels, thus protecting the brain from ischemic injury. Current research has shed a light on such a possibility. In fact, even physical exercise can modulate mitochondrial-mediated pathways and thus protect the brain from injury (Marques-Aleixo et al., 2012). Furthermore, a combination of various treatment strategies may be the most hopeful methodology due to flexibility and multiple targets of action.

However, more in-depth research is required for translatable and effective applications of these non-pharmaceutical approaches in clinical settings. For example, the benefits brought forth by the research and development of new devices alongside the advancement of physical therapies are limited and cannot be reliably quantified. As a result, therapies based on the pathophysiological, cellular and molecular biological mechanisms combined with the multimodal imaging are still in need of higher levels of clinical evidence. Moreover, although many studies show that acupuncture is potentially a useful modality for the treatment of stroke, its optimal conditions and therapeutic time windows for clinical practice need a strong basis supported by solid data from preclinical and translational research.

Translational medicine is the key success factor in developing stroke treatment, as well as improved the efficiency of translation. It is our belief that we may eventually find an efficient and effective way to apply non-pharmaceutical modalities to achieve maximal benefits for ischemic brain by scientifically quantifying therapeutic outcomes with reliable indexes and parameters, elucidating the precise mechanisms underlying the outcomes/effects, and eventually establishing practicable, and personalized therapeutic protocols.

Highlights.

  • Recent research in non-pharmaceutical therapies for ischemic stroke and potential mechanisms

  • Hypothermia, preconditioning, acupuncture, medical gas and other therapies for acute stroke

  • Mechanical endovascular recanalization and recovery devices for the chronic phase of stroke

Acknowledgments

This work was supported by the Ministry of Science and Technology, China (2011CB707804; 2012CB518502) and National Institute of Health, USA (AT-004422 and HD-034852).

Abbreviations

ACP

antegrade cerebral perfusion

AMES

assisted movement with enhanced sensation

AMPA

α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid

BAIPC

bilateral arm ischemic preconditioning

BBB

blood brain barrier

BCI

Brain Computer Interface

Bcl-2

B-cell lymphoma-2

BDNF

brain derived neurotrophic factor

CBF

cerebral blood flow

CREB

cyclic adenosine monophosphate response element binding protein

COOL AID

Cooling for Acute Ischaemic Brain Damage

COX-2

cyclooxygenase-2

DADLE

(D-Ala2, D-Leu5)-enkephalin)

DOR

δ-opioid receptor

DWI

diffusion-weighted imaging

EA

electro-acupuncture

eNOS

endothelial nitric oxide synthase

ERK

extracellular regulated protein kinases

ENT1

equilibrative nucleoside transporyer 1

FDA

Food and Drug Administration

FES

Functional Electrical Stimulation

GABA

gamma-aminobutyric acid

GluR2

glutamate receptor 2

GSK3β

Glycogen synthase kinase-3β

HBO

hyperbaric oxygen therapy

HIF

hypoxia inducible factor

HPC

Hypoxic preconditioning

HSP70

heat shock 70 kDa protein

IAS

intracranial arterial stenosis

ICTuS-L

Intravenous Thrombolysis Plus Hypothermia for Acute Treatment of Ischemic Stroke

IMS III

interventional management of Stroke III

IκB

inhibitor of NF-κB

IKK

inhibitor of NF-κB kinase

iNOS

inducible nitric oxide synthase

IPC

ischemic postconditioning

MAPK

mitogen-activated protein kinase

MCAO

middle cerebral artery occlusion

MMP

matrix metalloproteinase

MR RESCUE

mechanical retrieval and recanalization of stroke clots using embolectomy

mRS

modified Rankin Scale

NADPH

nicotinamide adenine dinucleotide phosphate

NBO

normobarichyperoxia therapy

NEST

NeuroThera Effectiveness and Safety Trial

nNOS

neuronal nitric oxide synthase

NF-κB

nuclear factor-κB

NIHSS

National Institutes of Health Stroke Scale

NMDA

N-methyl-D-aspartate

NOx

nitrite plus nitrate

PI3-kinase

phosphatidylinositol 3-kinase

PGC-1α

αsubunit of peroxisome proliferators-activated receptor-γcoactivator-1

PKA

protein kinase A

PKC

protein kinase C

pO2

partial oxygen

PPARγ

peroxisome proliferator-activated receptor γ

PTEN

phosphatase and tensin homologue

RIPostC

Limb remote ischemic postconditioning

RNS

reactive nitrogen species

ROS

reactive oxygen species

ROSC

Return of Spontaneous Circulation

rTMS

repetitive Transcranial Magnetic Stimulation

rtPA

recombinant tissue plasminogen activator

SphK

Sphingosine kinase

SPOTRIAS

specialized program in acute stroke

STAIR

Stroke Therapy Academic Industry Roundtable

TCM

Traditional Chinese Medicine

tDCS

transcranial Direct Current Stimulation

TIA

transient ischemic attack

TLT

Transcranial laser therapy

TNF-α

tumor necrosis factor α

VE

vesselendothelium

VNS

Vagus Nerve Stimulation

WHO

World Health Organization

Footnotes

Conflict of interest

All authors have no actual or potential conflict of interest including any financial, personal or other relationships with other people or organizations within three years of beginning of the submitted work.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Albert-Weissenberger C, Siren AL, Kleinschnitz C. Ischemic stroke and traumatic brain injury: the role of the kallikrein-kinin system. Progress in neurobiology. 2013;101–102:65–82. doi: 10.1016/j.pneurobio.2012.11.004. [DOI] [PubMed] [Google Scholar]
  2. Alexander DN, Cen S, Sullivan KJ, Bhavnani G, Ma X, Azen SP. Effects of acupuncture treatment on poststroke motor recovery and physical function: a pilot study. Neurorehabilitation and neural repair. 2004;18:259–267. doi: 10.1177/1545968304271568. [DOI] [PubMed] [Google Scholar]
  3. Allen CL, Bayraktutan U. Oxidative stress and its role in the pathogenesis of ischaemic stroke. International journal of stroke. 2009;4:461–470. doi: 10.1111/j.1747-4949.2009.00387.x. [DOI] [PubMed] [Google Scholar]
  4. Anderson DC, Bottini AG, Jagiella WM, Westphal B, Ford S, Rockswold GL, Loewenson RB. A pilot study of hyperbaric oxygen in the treatment of human stroke. Stroke. 1991;22:1137–1142. doi: 10.1161/01.str.22.9.1137. [DOI] [PubMed] [Google Scholar]
  5. Arai K, Lok J, Guo S, Hayakawa K, Xing C, Lo EH. Cellular mechanisms of neurovascular damage and repair after stroke. Journal of child neurology. 2011;26:1193–1198. doi: 10.1177/0883073811408610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Asakawa T, Xia Y. Future research in acupuncture—Better design and analysis for novel and valid findings. In: Xia, et al., editors. Current Research in Acupuncture. Springer; New York, Heidelberg, Dordrecht, London: 2012. pp. 687–726. [Google Scholar]
  7. Bai HM, Zhang YL, He JW, Zhang LC, Yang Y. Observation on therapeutic effect of comprehensive therapy for acute large area cerebral infarction. Chinese acupuncture & moxibustion. 2008;28:98–100. [PubMed] [Google Scholar]
  8. Balduini W, Carloni S, Buonocore G. Autophagy in hypoxia-ischemia induced brain injury. The journal of maternal-fetal & neonatal medicine. 2012;25(Suppl 1):30–34. doi: 10.3109/14767058.2012.663176. [DOI] [PubMed] [Google Scholar]
  9. Barach AL. Rare Gases Not Essential to Life. Science. 1934;80:593–594. doi: 10.1126/science.80.2086.593-a. [DOI] [PubMed] [Google Scholar]
  10. Bastani A, Jaberzadeh S. Does anodal transcranial direct current stimulation enhance excitability of the motor cortex and motor function in healthy individuals and subjects with stroke: a systematic review and meta-analysis. Clinical neurophysiology. 2012;123:644–657. doi: 10.1016/j.clinph.2011.08.029. [DOI] [PubMed] [Google Scholar]
  11. Behrens PF, Ostertag CB, Warnke PC. Regional cerebral blood flow in peritumoral brain edema during dexamethasone treatment: a xenon-enhanced computed tomographic study. Neurosurgery. 1998;43:235–240. doi: 10.1097/00006123-199808000-00027. discussion 240–231. [DOI] [PubMed] [Google Scholar]
  12. Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G, Smith K. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. The New England journal of medicine. 2002;346:557–563. doi: 10.1056/NEJMoa003289. [DOI] [PubMed] [Google Scholar]
  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 research. 2007;1152:228–233. doi: 10.1016/j.brainres.2007.03.068. [DOI] [PubMed] [Google Scholar]
  14. Borlongan CV, Hayashi T, Oeltgen PR, Su TP, Wang Y. Hibernation-like state induced by an opioid peptide protects against experimental stroke. BMC biology. 2009;7:31. doi: 10.1186/1741-7007-7-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Briyal S, Gulati A. Endothelin-A receptor antagonist BQ123 potentiates acetaminophen induced hypothermia and reduces infarction following focal cerebral ischemia in rats. European journal of pharmacology. 2010;644:73–79. doi: 10.1016/j.ejphar.2010.06.071. [DOI] [PubMed] [Google Scholar]
  16. Broderick JP, Palesch YY, Demchuk AM, Yeatts SD, Khatri P, Hill MD, Jauch EC, Jovin TG, Yan B, Silver FL, von Kummer R, Molina CA, Demaerschalk BM, Budzik R, Clark WM, Zaidat OO, Malisch TW, Goyal M, Schonewille WJ, Mazighi M, Engelter ST, Anderson C, Spilker J, Carrozzella J, Ryckborst KJ, Janis LS, Martin RH, Foster LD, Tomsick TA. Endovascular therapy after intravenous t-PA versus t-PA alone for stroke. The New England journal of medicine. 2013;368:893–903. doi: 10.1056/NEJMoa1214300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Busto R, Dietrich WD, Globus MY, Valdes I, Scheinberg P, Ginsberg MD. Small differences in intraischemic brain temperature critically determine the extent of ischemic neuronal injury. Journal of cerebral blood flow and metabolism. 1987;7:729–738. doi: 10.1038/jcbfm.1987.127. [DOI] [PubMed] [Google Scholar]
  18. Cai J, Kang Z, Liu WW, Luo X, Qiang S, Zhang JH, Ohta S, Sun X, Xu W, Tao H, Li R. Hydrogen therapy reduces apoptosis in neonatal hypoxia-ischemia rat model. Neuroscience letters. 2008;441:167–172. doi: 10.1016/j.neulet.2008.05.077. [DOI] [PubMed] [Google Scholar]
  19. Canevari L, Console A, Tendi EA, Clark JB, Bates TE. Effect of postischaemic hypothermia on the mitochondrial damage induced by ischaemia and reperfusion in the gerbil. Brain research. 1999;817:241–245. doi: 10.1016/s0006-8993(98)01278-5. [DOI] [PubMed] [Google Scholar]
  20. Carson S, McDonagh M, Russman B, Helfand M. Hyperbaric oxygen therapy for stroke: a systematic review of the evidence. Clinical rehabilitation. 2005;19:819–833. doi: 10.1191/0269215505cr907oa. [DOI] [PubMed] [Google Scholar]
  21. Castren M, Nordberg P, Svensson L, Taccone F, Vincent JL, Desruelles D, Eichwede F, Mols P, Schwab T, Vergnion M, Storm C, Pesenti A, Pachl J, Guerisse F, Elste T, Roessler M, Fritz H, Durnez P, Busch HJ, Inderbitzen B, Barbut D. Intra-arrest transnasal evaporative cooling: a randomized, prehospital, multicenter study (PRINCE: Pre-ROSC IntraNasal Cooling Effectiveness) Circulation. 2010;122:729–736. doi: 10.1161/CIRCULATIONAHA.109.931691. [DOI] [PubMed] [Google Scholar]
  22. Chao D, Bazzy-Asaad A, Balboni G, Salvadori S, Xia Y. Activation of DOR attenuates anoxic K+ derangement via inhibition of Na+ entry in mouse cortex. Cereb Cortex. 2008;18:2217–2227. doi: 10.1093/cercor/bhm247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chao D, Bazzy-Asaad A, Balboni G, Xia Y. delta-, but not mu-, opioid receptor stabilizes K(+) homeostasis by reducing Ca(2+) influx in the cortex during acute hypoxia. Journal of cellular physiology. 2007a;212:60–67. doi: 10.1002/jcp.21000. [DOI] [PubMed] [Google Scholar]
  24. Chao D, Donnelly DF, Feng Y, Bazzy-Asaad A, Xia Y. Cortical delta-opioid receptors potentiate K+ homeostasis during anoxia and oxygen-glucose deprivation. Journal of cerebral blood flow and metabolism. 2007b;27:356–368. doi: 10.1038/sj.jcbfm.9600352. [DOI] [PubMed] [Google Scholar]
  25. Chao D, Xia Y. Ionic storm in hypoxic/ischemic stress: can opioid receptors subside it? Progress in neurobiology. 2010;90:439–470. doi: 10.1016/j.pneurobio.2009.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chao DM, He XZ, Yang YL, Bazzy-Asaad A, Lazarus LH, Balboni G, Kim DH, Xia Y. DOR activation inhibits anoxic/ischemic Na+ influx through Na+ channels via PKC mechanisms in the cortex. Exp Neurol. 2012;236:228–239. doi: 10.1016/j.expneurol.2012.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chaturvedi M, Kaczmarek L. MMP-9 Inhibition: a Therapeutic Strategy in Ischemic Stroke. Molecular neurobiology. 2013 Sep 12; doi: 10.1007/s12035-013-8538-z. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chau AC, Fai Cheung RT, Jiang X, Au-Yeung PK, Li LS. An fMRI study showing the effect of acupuncture in chronic stage stroke patients with aphasia. Journal of acupuncture and meridian studies. 2010;3:53–57. doi: 10.1016/S2005-2901(10)60009-X. [DOI] [PubMed] [Google Scholar]
  29. Chen CH, Manaenko A, Zhan Y, Liu WW, Ostrowki RP, Tang J, Zhang JH. Hydrogen gas reduced acute hyperglycemia-enhanced hemorrhagic transformation in a focal ischemia rat model. Neuroscience. 2010;169:402–414. doi: 10.1016/j.neuroscience.2010.04.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chen H, Chopp M, Welch KM. Effect of mild hyperthermia on the ischemic infarct volume after middle cerebral artery occlusion in the rat. Neurology. 1991;41:1133–1135. doi: 10.1212/wnl.41.7.1133. [DOI] [PubMed] [Google Scholar]
  31. Chen YH, Lin XW, Fang JQ. Apoplexy. In: Wu HG, Fang JQ, Wang QQ, editors. Contemporary Research of Acupuncture and Moxibustion Therapy on Stubborn Diseases. Shanghai: Publishing House of Shanghai Science and Technology; 2002. [Google Scholar]
  32. Cheng O, Ostrowski RP, Wu B, Liu W, Chen C, Zhang JH. Cyclooxygenase-2 mediates hyperbaric oxygen preconditioning in the rat model of transient global cerebral ischemia. Stroke. 2011;42:484–490. doi: 10.1161/STROKEAHA.110.604421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Choi JS, Park J, Suk K, Moon C, Park YK, Han HS. Mild Hypothermia Attenuates Intercellular Adhesion Molecule-1 Induction via Activation of Extracellular Signal-Regulated Kinase-1/2 in a Focal Cerebral Ischemia Model. Stroke research and treatment. 2011;2011:846716. doi: 10.4061/2011/846716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Ciccone A, Valvassori L, Nichelatti M, Sgoifo A, Ponzio M, Sterzi R, Boccardi E. Endovascular treatment for acute ischemic stroke. The New England journal of medicine. 2013;368:904–913. doi: 10.1056/NEJMoa1213701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Clark DL, Penner M, Wowk S, Orellana-Jordan I, Colbourne F. Treatments (12 and 48 h) with systemic and brain-selective hypothermia techniques after permanent focal cerebral ischemia in rat. Experimental neurology. 2009;220:391–399. doi: 10.1016/j.expneurol.2009.10.002. [DOI] [PubMed] [Google Scholar]
  36. Clarke JB, Sokoloff L. Circulation and energy metabolism of the brain. In: Siegel GJ, Agranoff BW, Albers RW, Fisher SK, Uhler MD, editors. Basic neurochemistry. 6. Philadelphia: LippincottRaven; 1999. pp. 637–669. [Google Scholar]
  37. Clifton GL, Coffey CS, Fourwinds S, Zygun D, Valadka A, Smith KR, Jr, Frisby ML, Bucholz RD, Wilde EA, Levin HS, Okonkwo DO. Early induction of hypothermia for evacuated intracranial hematomas: a post hoc analysis of two clinical trials. Journal of neurosurgery. 2012;117:714–720. doi: 10.3171/2012.6.JNS111690. [DOI] [PubMed] [Google Scholar]
  38. Clifton GL, Valadka A, Zygun D, Coffey CS, Drever P, Fourwinds S, Janis LS, Wilde E, Taylor P, Harshman K, Conley A, Puccio A, Levin HS, McCauley SR, Bucholz RD, Smith KR, Schmidt JH, Scott JN, Yonas H, Okonkwo DO. Very early hypothermia induction in patients with severe brain injury (the National Acute Brain Injury Study: Hypothermia II): a randomised trial. Lancet neurology. 2011;10:131–139. doi: 10.1016/S1474-4422(10)70300-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Coburn M, Maze M, Franks NP. The neuroprotective effects of xenon and helium in an in vitro model of traumatic brain injury. Critical care medicine. 2008;36:588–595. doi: 10.1097/01.CCM.0B013E3181611F8A6. [DOI] [PubMed] [Google Scholar]
  40. Colbourne F, Corbett D. Delayed postischemic hypothermia: a six month survival study using behavioral and histological assessments of neuroprotection. The Journal of neuroscience. 1995;15:7250–7260. doi: 10.1523/JNEUROSCI.15-11-07250.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Colbourne F, Corbett D, Zhao Z, Yang J, Buchan AM. Prolonged but delayed postischemic hypothermia: a long-term outcome study in the rat middle cerebral artery occlusion model. Journal of cerebral blood flow and metabolism. 2000;20:1702–1708. doi: 10.1097/00004647-200012000-00009. [DOI] [PubMed] [Google Scholar]
  42. Colbourne F, Grooms SY, Zukin RS, Buchan AM, Bennett MV. Hypothermia rescues hippocampal CA1 neurons and attenuates down-regulation of the AMPA receptor GluR2 subunit after forebrain ischemia. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:2906–2910. doi: 10.1073/pnas.2628027100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Cordo P, Lutsep H, Cordo L, Wright WG, Cacciatore T, Skoss R. Assisted movement with enhanced sensation (AMES): coupling motor and sensory to remediate motor deficits in chronic stroke patients. Neurorehabilitation and neural repair. 2009;23:67–77. doi: 10.1177/1545968308317437. [DOI] [PubMed] [Google Scholar]
  44. Cui M, Bai X, Li T, Chen F, Dong Q, Zhao Y, Liu X. Decreased extracellular adenosine levels lead to loss of hypoxia-induced neuroprotection after repeated episodes of exposure to hypoxia. PloS one. 2013;8:e57065. doi: 10.1371/journal.pone.0057065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Curry A, Guo M, Patel R, Liebelt B, Sprague S, Lai Q, Zwagerman N, Cao FX, Jimenez D, Ding Y. Exercise pre-conditioning reduces brain inflammation in stroke via tumor necrosis factor-alpha, extracellular signal-regulated kinase 1/2 and matrix metalloproteinase-9 activity. Neurological research. 2010;32:756–762. doi: 10.1179/174313209X459101. [DOI] [PubMed] [Google Scholar]
  46. D’Brot J, Ahmed T. Hyperoxia prevents hypoxia-induced bronchial hyperreactivity via a cyclooxygenase-independent mechanism. J Appl Physiol. 1991;70:740–747. doi: 10.1152/jappl.1991.70.2.740. [DOI] [PubMed] [Google Scholar]
  47. Daly JJ, Cheng R, Rogers J, Litinas K, Hrovat K, Dohring M. Feasibility of a new application of noninvasive Brain Computer Interface (BCI): a case study of training for recovery of volitional motor control after stroke. Journal of neurologic physical therapy: JNPT. 2009;33:203–211. doi: 10.1097/NPT.0b013e3181c1fc0b. [DOI] [PubMed] [Google Scholar]
  48. David HN, Haelewyn B, Degoulet M, Colomb DG, Jr, Risso JJ, Abraini JH. Ex vivo and in vivo neuroprotection induced by argon when given after an excitotoxic or ischemic insult. PloS one. 2012a;7:e30934. doi: 10.1371/journal.pone.0030934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. 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. 2012b;112:2068–2076. doi: 10.1152/japplphysiol.00092.2012. [DOI] [PubMed] [Google Scholar]
  50. David HN, Haelewyn B, Risso JJ, Colloc’h N, Abraini JH. Xenon is an inhibitor of tissue-plasminogen activator: adverse and beneficial effects in a rat model of thromboembolic stroke. Journal of cerebral blood flow and metabolism. 2010;30:718–728. doi: 10.1038/jcbfm.2009.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Davis W, Mahale S, Carranza A, Cox B, Hayes K, Jimenez D, Ding Y. Exercise pre-conditioning ameliorates blood-brain barrier dysfunction in stroke by enhancing basal lamina. Neurological research. 2007;29:382–387. doi: 10.1179/016164107X204701. [DOI] [PubMed] [Google Scholar]
  52. De Georgia MA, Krieger DW, Abou-Chebl A, Devlin TG, Jauss M, Davis SM, Koroshetz WJ, Rordorf G, Warach S. Cooling for Acute Ischemic Brain Damage (COOL AID): a feasibility trial of endovascular cooling. Neurology. 2004;63:312–317. doi: 10.1212/01.wnl.0000129840.66938.75. [DOI] [PubMed] [Google Scholar]
  53. Della-Morte D, Guadagni F, Palmirotta R, Ferroni P, Testa G, Cacciatore F, Abete P, Rengo F, Perez-Pinzon MA, Sacco RL, Rundek T. Genetics and genomics of ischemic tolerance: focus on cardiac and cerebral ischemic preconditioning. Pharmacogenomics. 2012;13:1741–1757. doi: 10.2217/pgs.12.157. [DOI] [PubMed] [Google Scholar]
  54. Della-Morte D, Raval AP, Dave KR, Lin HW, Perez-Pinzon MA. Post-ischemic activation of protein kinase C epsilon protects the hippocampus from cerebral ischemic injury via alterations in cerebral blood flow. Neuroscience letters. 2011;487:158–162. doi: 10.1016/j.neulet.2010.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Deng H, Han HS, Cheng D, Sun GH, Yenari MA. Mild hypothermia inhibits inflammation after experimental stroke and brain inflammation. Stroke. 2003;34:2495–2501. doi: 10.1161/01.STR.0000091269.67384.E7. [DOI] [PubMed] [Google Scholar]
  56. Desmet KD, Paz DA, Corry JJ, Eells JT, Wong-Riley MT, Henry MM, Buchmann EV, Connelly MP, Dovi JV, Liang HL, Henshel DS, Yeager RL, Millsap DS, Lim J, Gould LJ, Das R, Jett M, Hodgson BD, Margolis D, Whelan HT. Clinical and experimental applications of NIR-LED photobiomodulation. Photomedicine and laser surgery. 2006;24:121–128. doi: 10.1089/pho.2006.24.121. [DOI] [PubMed] [Google Scholar]
  57. Ding GH, Liu RQ, Chao DM, Xia Y. Neuronal responses to hypoxic stress in mouse cognitive cortex. Circulation Research. 2012;111:A338. [Google Scholar]
  58. Ding YH, Ding Y, Li J, Bessert DA, Rafols JA. Exercise pre-conditioning strengthens brain microvascular integrity in a rat stroke model. Neurological research. 2006;28:184–189. doi: 10.1179/016164106X98053. [DOI] [PubMed] [Google Scholar]
  59. Dingley J, Hobbs C, Ferguson J, Stone J, Thoresen M. Xenon/hypothermia neuroprotection regimes in spontaneously breathing neonatal rats after hypoxic-ischemic insult: the respiratory and sedative effects. Anesthesia and analgesia. 2008;106:916–923. doi: 10.1213/ane.0b013e3181618669. table of contents. [DOI] [PubMed] [Google Scholar]
  60. Doufas AG, Lin CM, Suleman MI, Liem EB, Lenhardt R, Morioka N, Akca O, Shah YM, Bjorksten AR, Sessler DI. Dexmedetomidine and meperidine additively reduce the shivering threshold in humans. Stroke. 2003;34:1218–1223. doi: 10.1161/01.STR.0000068787.76670.A4. [DOI] [PubMed] [Google Scholar]
  61. Du Y, Li C, Hu WW, Song YJ, Zhang GY. Neuroprotection of preconditioning against ischemic brain injury in rat hippocampus through inhibition of the assembly of GluR6-PSD95-mixed lineage kinase 3 signaling module via nuclear and non-nuclear pathways. Neuroscience. 2009;161:370–380. doi: 10.1016/j.neuroscience.2009.03.050. [DOI] [PubMed] [Google Scholar]
  62. Duz B, Oztas E, Erginay T, Erdogan E, Gonul E. The effect of moderate hypothermia in acute ischemic stroke on pericyte migration: an ultrastructural study. Cryobiology. 2007;55:279–284. doi: 10.1016/j.cryobiol.2007.08.009. [DOI] [PubMed] [Google Scholar]
  63. Erecinska M, Thoresen M, Silver IA. Effects of hypothermia on energy metabolism in Mammalian central nervous system. Journal of cerebral blood flow and metabolism. 2003;23:513–530. doi: 10.1097/01.WCB.0000066287.21705.21. [DOI] [PubMed] [Google Scholar]
  64. Fairbanks SL, Brambrink AM. Preconditioning and postconditioning for neuroprotection: the most recent evidence. Best practice & research. Clinical anaesthesiology. 2010;24:521–534. doi: 10.1016/j.bpa.2010.10.004. [DOI] [PubMed] [Google Scholar]
  65. Fairchild KD, Singh IS, Patel S, Drysdale BE, Viscardi RM, Hester L, Lazusky HM, Hasday JD. Hypothermia prolongs activation of NF-kappaB and augments generation of inflammatory cytokines. American journal of physiology. Cell physiology. 2004;287:C422–431. doi: 10.1152/ajpcell.00507.2003. [DOI] [PubMed] [Google Scholar]
  66. Fisher M, Feuerstein G, Howells DW, Hurn PD, Kent TA, Savitz SI, Lo EH. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke. 2009;40:2244–2250. doi: 10.1161/STROKEAHA.108.541128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Fisher M, Bastan B. Identifying and utilizing the ischemic penumbra. Neurology. 2012;79:S79–85. doi: 10.1212/WNL.0b013e3182695814. [DOI] [PubMed] [Google Scholar]
  68. Fries M, Brucken A, Cizen A, Westerkamp M, Lower C, Deike-Glindemann J, Schnorrenberger NK, Rex S, Coburn M, Nolte KW, Weis J, Rossaint R, Derwall M. Combining xenon and mild therapeutic hypothermia preserves neurological function after prolonged cardiac arrest in pigs. Critical care medicine. 2012;40:1297–1303. doi: 10.1097/CCM.0b013e31823c8ce7. [DOI] [PubMed] [Google Scholar]
  69. Gao X, Ren C, Zhao H. Protective effects of ischemic postconditioning compared with gradual reperfusion or preconditioning. Journal of neuroscience research. 2008a;86:2505–2511. doi: 10.1002/jnr.21703. [DOI] [PubMed] [Google Scholar]
  70. Gao X, Zhang H, Takahashi T, Hsieh J, Liao J, Steinberg GK, Zhao H. The Akt signaling pathway contributes to postconditioning’s protection against stroke; the protection is associated with the MAPK and PKC pathways. Journal of neurochemistry. 2008b;105:943–955. doi: 10.1111/j.1471-4159.2008.05218.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Gosman-Hedstrom G, Claesson L, Klingenstierna U, Carlsson J, Olausson B, Frizell M, Fagerberg B, Blomstrand C. Effects of acupuncture treatment on daily life activities and quality of life: a controlled, prospective, and randomized study of acute stroke patients. Stroke. 1998;29:2100–2108. doi: 10.1161/01.str.29.10.2100. [DOI] [PubMed] [Google Scholar]
  72. Gotberg M, van der Pals J, Olivecrona GK, Koul S, Erlinge D. Mild hypothermia reduces acute mortality and improves hemodynamic outcome in a cardiogenic shock pig model. Resuscitation. 2010;81:1190–1196. doi: 10.1016/j.resuscitation.2010.04.033. [DOI] [PubMed] [Google Scholar]
  73. Govindaswami M, Brown SA, Yu J, Zhu H, Bishop PD, Kindy MS, Oeltgen PR. Delta 2-specific opioid receptor agonist and hibernating woodchuck plasma fraction provide ischemic neuroprotection. Acad Emerg Med. 2008;15:250–257. doi: 10.1111/j.1553-2712.2008.00048.x. [DOI] [PubMed] [Google Scholar]
  74. Group, H. a. C. A. S. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. The New England journal of medicine. 2002;346:549–556. doi: 10.1056/NEJMoa012689. [DOI] [PubMed] [Google Scholar]
  75. Guo JC, Cheng JS, Xia Y. Acupuncture Therapy for Neurological Diseases: A Neurobiological View. Springer-Tsinghua Press; Heidelberg, London, New York, Beijing: 2010. Acupuncture therapy for stroke; pp. 226–262. [Google Scholar]
  76. Gutsaeva DR, Carraway MS, Suliman HB, Demchenko IT, Shitara H, Yonekawa H, Piantadosi CA. Transient hypoxia stimulates mitochondrial biogenesis in brain subcortex by a neuronal nitric oxide synthase-dependent mechanism. The Journal of neuroscience. 2008;28:2015–2024. doi: 10.1523/JNEUROSCI.5654-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hadjiev DI, Mineva PP. Normobaric hyperoxia treatment in acute ischemic stroke. Stroke. 2009;40:e636. doi: 10.1161/STROKEAHA.109.563155. author reply e637. [DOI] [PubMed] [Google Scholar]
  78. Hadjiev DI, Mineva PP. Transient ischemic attack may present a target for normobaric hyperoxia treatment. Medical hypotheses. 2010;75:128–130. doi: 10.1016/j.mehy.2010.02.008. [DOI] [PubMed] [Google Scholar]
  79. Hajat C, Hajat S, Sharma P. Effects of poststroke pyrexia on stroke outcome: a meta-analysis of studies in patients. Stroke. 2000;31:410–414. doi: 10.1161/01.str.31.2.410. [DOI] [PubMed] [Google Scholar]
  80. Han HS, Karabiyikoglu M, Kelly S, Sobel RA, Yenari MA. Mild hypothermia inhibits nuclear factor-kappaB translocation in experimental stroke. Journal of cerebral blood flow and metabolism. 2003;23:589–598. doi: 10.1097/01.WCB.0000059566.39780.8D. [DOI] [PubMed] [Google Scholar]
  81. Hansen PR, Thibault H, Abdulla J. Postconditioning during primary percutaneous coronary intervention: a review and meta-analysis. International journal of cardiology. 2010;144:22–25. doi: 10.1016/j.ijcard.2009.03.118. [DOI] [PubMed] [Google Scholar]
  82. Hasseldam H, Hansen-Schwartz J, Munkholm N, Hou J, Johansen FF. Remote post-conditioning reduces hypoxic damage early after experimental stroke. Neurological research. 2013;35:336–343. doi: 10.1179/1743132812Y.0000000130. [DOI] [PubMed] [Google Scholar]
  83. Hawkins BT, Egleton RD. Pathophysiology of the blood-brain barrier: animal models and methods. Current topics in developmental biology. 2008;80:277–309. doi: 10.1016/S0070-2153(07)80007-X. [DOI] [PubMed] [Google Scholar]
  84. He X, Sandhu HK, Yang Y, Hua F, Belser N, Kim DH, Xia Y. Neuroprotection against hypoxia/ischemia: delta-opioid receptor-mediated cellular/molecular events. Cellular and molecular life sciences. 2013;70:2291–2303. doi: 10.1007/s00018-012-1167-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. He X, Xu X, Fan M, Chen X, Sun X, Luo G, Chen L, Mu Q, Feng Y, Mao Q, Chao Z. Preconditioning with hyperbaric oxygen induces tolerance against renal ischemia-reperfusion injury via increased expression of heme oxygenase-1. The Journal of surgical research. 2011;170:e271–277. doi: 10.1016/j.jss.2011.06.008. [DOI] [PubMed] [Google Scholar]
  86. Hemmen TM, Raman R, Guluma KZ, Meyer BC, Gomes JA, Cruz-Flores S, Wijman CA, Rapp KS, Grotta JC, Lyden PD. Intravenous thrombolysis plus hypothermia for acute treatment of ischemic stroke (ICTuS-L): final results. Stroke. 2010;41:2265–2270. doi: 10.1161/STROKEAHA.110.592295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. 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. Journal of cerebral blood flow and metabolism. 2007;27:1632–1642. doi: 10.1038/sj.jcbfm.9600463. [DOI] [PubMed] [Google Scholar]
  88. Henninger N, Bratane BT, Bastan B, Bouley J, Fisher M. Normobaric hyperoxia and delayed tPA treatment in a rat embolic stroke model. Journal of cerebral blood flow and metabolism. 2009;29:119–129. doi: 10.1038/jcbfm.2008.104. [DOI] [PubMed] [Google Scholar]
  89. Henninger N, Fisher M. Normobaric hyperoxia - a promising approach to expand the time window for acute stroke treatment. Cerebrovasc Dis. 2006;21:134–136. doi: 10.1159/000090446. [DOI] [PubMed] [Google Scholar]
  90. Hess DC, Hoda MN, Bhatia K. Remote limb preconditioning and postconditioning: will it translate into a promising treatment for acute stroke? Stroke. 2013;44:1191–1197. doi: 10.1161/STROKEAHA.112.678482. [DOI] [PubMed] [Google Scholar]
  91. Hitchcock KE, Holland CK. Ultrasound-assisted thrombolysis for stroke therapy: better thrombus break-up with bubbles. Stroke. 2010;41:S50–53. doi: 10.1161/STROKEAHA.110.595348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Hong DM, Lee EH, Kim HJ, Min JJ, Chin JH, Choi DK, Bahk JH, Sim JY, Choi IC, Jeon Y. Does remote ischaemic preconditioning with postconditioning improve clinical outcomes of patients undergoing cardiac surgery? Remote Ischaemic Preconditioning with Postconditioning Outcome Trial. European heart journal. 2013 Sep 7; doi: 10.1093/eurheartj/eht346. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  93. Hong SS, Qian H, Zhao P, Bazzy-Asaad A, Xia Y. Anisomycin protects cortical neurons from prolonged hypoxia with differential regulation of p38 and ERK. Brain research. 2007;1149:76–86. doi: 10.1016/j.brainres.2007.02.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Hopwood V, Lewith G, Prescott P, Campbell MJ. Evaluating the efficacy of acupuncture in defined aspects of stroke recovery: a randomised, placebo controlled single blind study. Journal of neurology. 2008;255:858–866. doi: 10.1007/s00415-008-0790-1. [DOI] [PubMed] [Google Scholar]
  95. Hu D, Chen F, Guan C, Yang F, Qu Y. Anti-hypoxia effect of adenovirus-mediated expression of heat shock protein 70 (HSP70) on primary cultured neurons. Journal of neuroscience research. 2013;91:1174–1182. doi: 10.1002/jnr.23240. [DOI] [PubMed] [Google Scholar]
  96. Hu HH, Chung C, Liu TJ, Chen RC, Chen CH, Chou P, Huang WS, Lin JC, Tsuei JJ. A randomized controlled trial on the treatment for acute partial ischemic stroke with acupuncture. Neuroepidemiology. 1993;12:106–113. doi: 10.1159/000110308. [DOI] [PubMed] [Google Scholar]
  97. Iwata M, Inoue S, Kawaguchi M, Kurita N, Horiuchi T, Nakamura M, Konishi N, Furuya H. Delta opioid receptors stimulation with [D-Ala2, D-Leu5] enkephalin does not provide neuroprotection in the hippocampus in rats subjected to forebrain ischemia. Neuroscience letters. 2007;414:242–246. doi: 10.1016/j.neulet.2006.12.021. [DOI] [PubMed] [Google Scholar]
  98. Jauch EC, Saver JL, Adams HP, Jr, Bruno A, Connors JJ, Demaerschalk BM, Khatri P, McMullan PW, Jr, Qureshi AI, Rosenfield K, Scott PA, Summers DR, Wang DZ, Wintermark M, Yonas H. Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2013;44:870–947. doi: 10.1161/STR.0b013e318284056a. [DOI] [PubMed] [Google Scholar]
  99. Jian Liu K, Rosenberg GA. Matrix metalloproteinases and free radicals in cerebral ischemia. Free radical biology & medicine. 2005;39:71–80. doi: 10.1016/j.freeradbiomed.2005.03.033. [DOI] [PubMed] [Google Scholar]
  100. 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. Experimental neurology. 2013;240:9–16. doi: 10.1016/j.expneurol.2012.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Johansson K, Lindgren I, Widner H, Wiklund I, Johansson BB. Can sensory stimulation improve the functional outcome in stroke patients? Neurology. 1993;43:2189–2192. doi: 10.1212/wnl.43.11.2189. [DOI] [PubMed] [Google Scholar]
  102. Kallmunzer B, Schwab S, Kollmar R. Mild hypothermia of 34 degrees C reduces side effects of rt-PA treatment after thromboembolic stroke in rats. Experimental & translational stroke medicine. 2012;4:3. doi: 10.1186/2040-7378-4-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Kammersgaard LP, Rasmussen BH, Jorgensen HS, Reith J, Weber U, Olsen TS. Feasibility and safety of inducing modest hypothermia in awake patients with acute stroke through surface cooling: A case-control study: the Copenhagen Stroke Study. Stroke. 2000;31:2251–2256. doi: 10.1161/01.str.31.9.2251. [DOI] [PubMed] [Google Scholar]
  104. Kaneko T, Kibayashi K. Mild hypothermia facilitates the expression of cold-inducible RNA-binding protein and heat shock protein 70.1 in mouse brain. Brain research. 2012;1466:128–136. doi: 10.1016/j.brainres.2012.05.001. [DOI] [PubMed] [Google Scholar]
  105. Kang X, Chao D, Gu Q, Ding G, Wang Y, Balboni G, Lazarus LH, Xia Y. delta-Opioid receptors protect from anoxic disruption of Na+ homeostasis via Na+ channel regulation. Cellular and molecular life sciences. 2009;66:3505–3516. doi: 10.1007/s00018-009-0136-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Kheir JN, Scharp LA, Borden MA, Swanson EJ, Loxley A, Reese JH, Black KJ, Velazquez LA, Thomson LM, Walsh BK, Mullen KE, Graham DA, Lawlor MW, Brugnara C, Bell DC, McGowan FX., Jr Oxygen gas-filled microparticles provide intravenous oxygen delivery. Science translational medicine. 2012;4:140ra188. doi: 10.1126/scitranslmed.3003679. [DOI] [PubMed] [Google Scholar]
  107. Khodaparast N, Hays SA, Sloan AM, Hulsey DR, Ruiz A, Pantoja M, Rennaker RL, 2nd, Kilgard MP. Vagus nerve stimulation during rehabilitative training improves forelimb strength following ischemic stroke. Neurobiology of disease. 2013;60C:80–88. doi: 10.1016/j.nbd.2013.08.002. [DOI] [PubMed] [Google Scholar]
  108. Kidwell CS, Jahan R, Gornbein J, Alger JR, Nenov V, Ajani Z, Feng L, Meyer BC, Olson S, Schwamm LH, Yoo AJ, Marshall RS, Meyers PM, Yavagal DR, Wintermark M, Guzy J, Starkman S, Saver JL. A trial of imaging selection and endovascular treatment for ischemic stroke. The New England journal of medicine. 2013;368:914–923. doi: 10.1056/NEJMoa1212793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Kim EJ, Raval AP, Hirsch N, Perez-Pinzon MA. Ischemic Preconditioning Mediates Cyclooxygenase-2 Expression Via Nuclear Factor-Kappa B Activation in Mixed Cortical Neuronal Cultures. Translational stroke research. 2010;1:40–47. doi: 10.1007/s12975-009-0006-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Kim HY, Singhal AB, Lo EH. Normobaric hyperoxia extends the reperfusion window in focal cerebral ischemia. Annals of neurology. 2005;57:571–575. doi: 10.1002/ana.20430. [DOI] [PubMed] [Google Scholar]
  111. Kitagawa K, Matsumoto M, Kuwabara K, Tagaya M, Ohtsuki T, Hata R, Ueda H, Handa N, Kimura K, Kamada T. ‘Ischemic tolerance’ phenomenon detected in various brain regions. Brain research. 1991;561:203–211. doi: 10.1016/0006-8993(91)91596-s. [DOI] [PubMed] [Google Scholar]
  112. Koch S, Sacco RL, Perez-Pinzon MA. Preconditioning the brain: moving on to the next frontier of neurotherapeutics. Stroke. 2012;43:1455–1457. doi: 10.1161/STROKEAHA.111.646919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Kondo T, Kakuda W, Yamada N, Shimizu M, Hagino H, Abo M. Effect of low-frequency rTMS on motor neuron excitability after stroke. Acta neurologica Scandinavica. 2013;127:26–30. doi: 10.1111/j.1600-0404.2012.01669.x. [DOI] [PubMed] [Google Scholar]
  114. Kong JC, Lee MS, Shin BC, Song YS, Ernst E. Acupuncture for functional recovery after stroke: a systematic review of sham-controlled randomized clinical trials. Canadian Medical Association journal. 2010;182:1723–1729. doi: 10.1503/cmaj.091113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Konstantinov IE, Arab S, Kharbanda RK, Li J, Cheung MM, Cherepanov V, Downey GP, Liu PP, Cukerman E, Coles JG, Redington AN. The remote ischemic preconditioning stimulus modifies inflammatory gene expression in humans. Physiological genomics. 2004;19:143–150. doi: 10.1152/physiolgenomics.00046.2004. [DOI] [PubMed] [Google Scholar]
  116. Krieger DW, De Georgia MA, Abou-Chebl A, Andrefsky JC, Sila CA, Katzan IL, Mayberg MR, Furlan AJ. Cooling for acute ischemic brain damage (cool aid): an open pilot study of induced hypothermia in acute ischemic stroke. Stroke. 2001;32:1847–1854. doi: 10.1161/01.str.32.8.1847. [DOI] [PubMed] [Google Scholar]
  117. Lakhan SE, Pamplona F. Application of mild therapeutic hypothermia on stroke: a systematic review and meta-analysis. Stroke research and treatment. 2012;2012:295906. doi: 10.1155/2012/295906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Lampl Y, Zivin JA, Fisher M, Lew R, Welin L, Dahlof B, Borenstein P, Andersson B, Perez J, Caparo C, Ilic S, Oron U. Infrared laser therapy for ischemic stroke: a new treatment strategy: results of the NeuroThera Effectiveness and Safety Trial-1 (NEST-1) Stroke. 2007;38:1843–1849. doi: 10.1161/STROKEAHA.106.478230. [DOI] [PubMed] [Google Scholar]
  119. Lapchak PA. Taking a light approach to treating acute ischemic stroke patients: transcranial near-infrared laser therapy translational science. Annals of medicine. 2010;42:576–586. doi: 10.3109/07853890.2010.532811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Lapchak PA, De Taboada L. Transcranial near infrared laser treatment (NILT) increases cortical adenosine-5′-triphosphate (ATP) content following embolic strokes in rabbits. Brain research. 2010;1306:100–105. doi: 10.1016/j.brainres.2009.10.022. [DOI] [PubMed] [Google Scholar]
  121. Lapchak PA, Han MK, Salgado KF, Streeter J, Zivin JA. Safety profile of transcranial near-infrared laser therapy administered in combination with thrombolytic therapy to embolized rabbits. Stroke. 2008;39:3073–3078. doi: 10.1161/STROKEAHA.108.516393. [DOI] [PubMed] [Google Scholar]
  122. Lee JE, Yoon YJ, Moseley ME, Yenari MA. Reduction in levels of matrix metalloproteinases and increased expression of tissue inhibitor of metalloproteinase-2 in response to mild hypothermia therapy in experimental stroke. Journal of neurosurgery. 2005;103:289–297. doi: 10.3171/jns.2005.103.2.0289. [DOI] [PubMed] [Google Scholar]
  123. Lee SM, Zhao H, Maier CM, Steinberg GK. The protective effect of early hypothermia on PTEN phosphorylation correlates with free radical inhibition in rat stroke. Journal of cerebral blood flow and metabolism. 2009;29:1589–1600. doi: 10.1038/jcbfm.2009.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Leung MC, Lo SC, Siu FK, So KF. Treatment of experimentally induced transient cerebral ischemia with low energy laser inhibits nitric oxide synthase activity and up-regulates the expression of transforming growth factor-beta 1. Lasers in surgery and medicine. 2002;31:283–288. doi: 10.1002/lsm.10096. [DOI] [PubMed] [Google Scholar]
  125. Li J, Luan X, Clark JC, Rafols JA, Ding Y. Neuroprotection against transient cerebral ischemia by exercise pre-conditioning in rats. Neurological research. 2004;26:404–408. doi: 10.1179/016164104225016038. [DOI] [PubMed] [Google Scholar]
  126. Li JS, Zhang W, Kang ZM, Ding SJ, Liu WW, Zhang JH, Guan YT, Sun XJ. Hyperbaric oxygen preconditioning reduces ischemia-reperfusion injury by inhibition of apoptosis via mitochondrial pathway in rat brain. Neuroscience. 2009a;159:1309–1315. doi: 10.1016/j.neuroscience.2009.01.011. [DOI] [PubMed] [Google Scholar]
  127. Li M, Sun JH, Lu JW, Liu LY, Geng H. Observation on theraputic effect of five-needle-in-nape acupuncture for treatment of poststroke pesudobulbar paralysis dysphagia. Chinese acupuncture & moxibustion. 2009b;29:873–875. [PubMed] [Google Scholar]
  128. Li P, Longhurst JC. Neural mechanism of electroacupuncture’s hypotensive effects. Autonomic neuroscience: basic & clinical. 2010;157:24–30. doi: 10.1016/j.autneu.2010.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Li X, Luo P, Wang Q, Xiong L. Electroacupuncture Pretreatment as a Novel Avenue to Protect Brain against Ischemia and Reperfusion Injury. Evid Based Complement Alternat Med. 2012:195–397. doi: 10.1155/2012/195397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Liang JF, Xia Y. Acupuncture modulation of neural transmitters/modulators. In: Xia, et al., editors. Current Research in Acupuncture. Springer; New York, Heidelberg, Dordrecht, London: 2012. pp. 1–36. [Google Scholar]
  131. Liebner S, Czupalla CJ, Wolburg H. Current concepts of blood-brain barrier development. The International journal of developmental biology. 2011;55:467–476. doi: 10.1387/ijdb.103224sl. [DOI] [PubMed] [Google Scholar]
  132. Lim SY, Hausenloy DJ. Remote ischemic conditioning: from bench to bedside. Frontiers in physiology. 2012;3:27. doi: 10.3389/fphys.2012.00027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Lim YJ, Zheng S, Zuo Z. Morphine preconditions Purkinje cells against cell death under in vitro simulated ischemia-reperfusion conditions. Anesthesiology. 2004;100:562–568. doi: 10.1097/00000542-200403000-00015. [DOI] [PubMed] [Google Scholar]
  134. Lin HW, Thompson JW, Morris KC, Perez-Pinzon MA. Signal transducers and activators of transcription: STATs-mediated mitochondrial neuroprotection. Antioxidants & redox signaling. 2011;14:1853–1861. doi: 10.1089/ars.2010.3467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Liu C, Weaver J, Liu KJ. Rapid conditioning with oxygen oscillation: neuroprotection by intermittent normobaric hyperoxia after transient focal cerebral ischemia in rats. Stroke. 2012a;43:220–226. doi: 10.1161/STROKEAHA.111.625756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Liu L, Kim JY, Koike MA, Yoon YJ, Tang XN, Ma H, Lee H, Steinberg GK, Lee JE, Yenari MA. FasL shedding is reduced by hypothermia in experimental stroke. Journal of neurochemistry. 2008a;106:541–550. doi: 10.1111/j.1471-4159.2008.05411.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Liu M. Acupuncture for stroke in China: needing more high-quality evidence. International journal of stroke. 2006a;1:34–35. doi: 10.1111/j.1747-4949.2005.00003.x. [DOI] [PubMed] [Google Scholar]
  138. 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. Journal of cerebral blood flow and metabolism. 2006b;26:1274–1284. doi: 10.1038/sj.jcbfm.9600277. [DOI] [PubMed] [Google Scholar]
  139. 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. Journal of cerebral blood flow and metabolism. 2004;24:343–349. doi: 10.1097/01.WCB.0000110047.43905.01. [DOI] [PubMed] [Google Scholar]
  140. Liu SY, Hsieh CL, Wei TS, Liu PT, Chang YJ, Li TC. Acupuncture stimulation improves balance function in stroke patients: a single-blinded controlled, randomized study. The American journal of Chinese medicine. 2009a;37:483–494. doi: 10.1142/S0192415X09006990. [DOI] [PubMed] [Google Scholar]
  141. Liu W, Chen Q, Liu J, Liu KJ. Normobaric hyperoxia protects the blood brain barrier through inhibiting Nox2 containing NADPH oxidase in ischemic stroke. Medical gas research. 2011a;1:22. doi: 10.1186/2045-9912-1-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. 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. 2009b;40:2526–2531. doi: 10.1161/STROKEAHA.108.545483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. 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. Journal of neurochemistry. 2009c;108:811–820. doi: 10.1111/j.1471-4159.2008.05821.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Liu W, Khatibi N, Sridharan A, Zhang JH. Application of medical gases in the field of neurobiology. Medical gas research. 2011b;1:13. doi: 10.1186/2045-9912-1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Liu W, Liu K, Tao H, Chen C, Zhang JH, Sun X. Hyperoxia preconditioning: the next frontier in neurology? Neurological research. 2012b;34:415–421. doi: 10.1179/1743132812Y.0000000034. [DOI] [PubMed] [Google Scholar]
  146. Liu W, Mukherjee M, Sun C, Liu H, McPeak LK. Electroacupuncture may help motor recovery in chronic stroke survivors: a pilot study. Journal of rehabilitation research and development. 2008b;45:587–595. doi: 10.1682/jrrd.2007.11.0181. [DOI] [PubMed] [Google Scholar]
  147. Liu W, Sood R, Chen Q, Sakoglu U, Hendren J, Cetin O, Miyake M, Liu KJ. Normobaric hyperoxia inhibits NADPH oxidase-mediated matrix metalloproteinase-9 induction in cerebral microvessels in experimental stroke. Journal of neurochemistry. 2008c;107:1196–1205. doi: 10.1111/j.1471-4159.2008.05664.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Liu XR, Luo M, Yan F, Zhang CC, Li SJ, Zhao HP, Ji XM, Luo YM. Ischemic postconditioning diminishes matrix metalloproteinase 9 expression and attenuates loss of the extracellular matrix proteins in rats following middle cerebral artery occlusion and reperfusion. CNS neuroscience & therapeutics. 2012c;18:855–863. doi: 10.1111/j.1755-5949.2012.00366.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Liu Y, Sun XJ, Liu J, Kang ZM, Deng XM. Heme oxygenase-1 could mediate the protective effects of hyperbaric oxygen preconditioning against hepatic ischemia-reperfusion injury in rats. Clinical and experimental pharmacology & physiology. 2011c;38:675–682. doi: 10.1111/j.1440-1681.2011.05560.x. [DOI] [PubMed] [Google Scholar]
  150. Loetscher PD, Rossaint J, Rossaint R, Weis J, Fries M, Fahlenkamp A, Ryang YM, Grottke O, Coburn M. Argon: neuroprotection in in vitro models of cerebral ischemia and traumatic brain injury. Crit Care. 2009;13:R206. doi: 10.1186/cc8214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Ma D, Lim T, Xu J, Tang H, Wan Y, Zhao H, Hossain M, Maxwell PH, Maze M. Xenon preconditioning protects against renal ischemic-reperfusion injury via HIF-1alpha activation. Journal of the American Society of Nephrology. 2009;20:713–720. doi: 10.1681/ASN.2008070712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Ma MC, Qian H, Ghassemi F, Zhao P, Xia Y. Oxygen-sensitive delta-opioid receptor-regulated survival and death signals: novel insights into neuronal preconditioning and protection. The Journal of biological chemistry. 2005;280:16208–16218. doi: 10.1074/jbc.M408055200. [DOI] [PubMed] [Google Scholar]
  153. Mariak Z, White MD, Lewko J, Lyson T, Piekarski P. Direct cooling of the human brain by heat loss from the upper respiratory tract. J Appl Physiol. 1999;87:1609–1613. doi: 10.1152/jappl.1999.87.5.1609. [DOI] [PubMed] [Google Scholar]
  154. Marques-Aleixo I, Oliveira PJ, Moreira PI, Magalhaes J, Ascensao A. Physical exercise as a possible strategy for brain protection: evidence from mitochondrial-mediated mechanisms. Progress in neurobiology. 2012;99:149–162. doi: 10.1016/j.pneurobio.2012.08.002. [DOI] [PubMed] [Google Scholar]
  155. Matchett GA, Fathali N, Hasegawa Y, Jadhav V, Ostrowski RP, Martin RD, Dorotta IR, Sun X, Zhang JH. Hydrogen gas is ineffective in moderate and severe neonatal hypoxia-ischemia rat models. Brain research. 2009;1259:90–97. doi: 10.1016/j.brainres.2008.12.066. [DOI] [PubMed] [Google Scholar]
  156. Matsui T, Kakeda T. IL-10 production is reduced by hypothermia but augmented by hyperthermia in rat microglia. Journal of neurotrauma. 2008;25:709–715. doi: 10.1089/neu.2007.0482. [DOI] [PubMed] [Google Scholar]
  157. McCauley SR, Wilde EA, Moretti P, Macleod MC, Pedroza C, Drever P, Fourwinds S, Frisby ML, Beers SR, Scott JN, Hunter JV, Traipe E, Valadka AB, Okonkwo DO, Zygun DA, Puccio AM, Clifton GL. Neurological outcome scale for traumatic brain injury: III. Criterion-related validity and sensitivity to change in the NABIS hypothermia-II clinical trial. Journal of neurotrauma. 2013;30:1506–1511. doi: 10.1089/neu.2013.2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Meng R, Asmaro K, Meng L, Liu Y, Ma C, Xi C, Li G, Ren C, Luo Y, Ling F, Jia J, Hua Y, Wang X, Ding Y, Lo EH, Ji X. Upper limb ischemic preconditioning prevents recurrent stroke in intracranial arterial stenosis. Neurology. 2012;79:1853–1861. doi: 10.1212/WNL.0b013e318271f76a. [DOI] [PubMed] [Google Scholar]
  159. Mochizuki-Oda N, Kataoka Y, Cui Y, Yamada H, Heya M, Awazu K. Effects of near-infra-red laser irradiation on adenosine triphosphate and adenosine diphosphate contents of rat brain tissue. Neuroscience letters. 2002;323:207–210. doi: 10.1016/s0304-3940(02)00159-3. [DOI] [PubMed] [Google Scholar]
  160. Moncayo J, de Freitas GR, Bogousslavsky J, Altieri M, van Melle G. Do transient ischemic attacks have a neuroprotective effect? Neurology. 2000;54:2089–2094. doi: 10.1212/wnl.54.11.2089. [DOI] [PubMed] [Google Scholar]
  161. Moon SK, Whang YK, Park SU, Ko CN, Kim YS, Bae HS, Cho KH. Antispastic effect of electroacupuncture and moxibustion in stroke patients. The American journal of Chinese medicine. 2003;31:467–474. doi: 10.1142/S0192415X03001077. [DOI] [PubMed] [Google Scholar]
  162. Morris KC, Lin HW, Thompson JW, Perez-Pinzon MA. Pathways for ischemic cytoprotection: role of sirtuins in caloric restriction, resveratrol, and ischemic preconditioning. Journal of cerebral blood flow and metabolism. 2011;31:1003–1019. doi: 10.1038/jcbfm.2010.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124–1136. doi: 10.1161/01.cir.74.5.1124. [DOI] [PubMed] [Google Scholar]
  164. Naeser MA, Alexander MP, Stiassny-Eder D, Galler V, Hobbs J, Bachman D. Acupuncture in the treatment of paralysis in chronic and acute stroke patients--improvement correlated with specific CT scan lesion sites. Acupuncture & electro-therapeutics research. 1994;19:227–249. doi: 10.3727/036012994816357231. [DOI] [PubMed] [Google Scholar]
  165. Narayanan SV, Dave KR, Perez-Pinzon MA. Ischemic preconditioning and clinical scenarios. Current opinion in neurology. 2013;26:1–7. doi: 10.1097/WCO.0b013e32835bf200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Natale G, Cattano D, Abramo A, Forfori F, Fulceri F, Fornai F, Paparelli A, Giunta F. Morphological evidence that xenon neuroprotects against N-methyl-DL-aspartic acid-induced damage in the rat arcuate nucleus: a time-dependent study. Annals of the New York Academy of Sciences. 2006;1074:650–658. doi: 10.1196/annals.1369.063. [DOI] [PubMed] [Google Scholar]
  167. Nighoghossian N, Trouillas P, Adeleine P, Salord F. Hyperbaric oxygen in the treatment of acute ischemic stroke. A double-blind pilot study. Stroke. 1995;26:1369–1372. doi: 10.1161/01.str.26.8.1369. [DOI] [PubMed] [Google Scholar]
  168. Nogueira RG, Lutsep HL, Gupta R, Jovin TG, Albers GW, Walker GA, Liebeskind DS, Smith WS. Trevo versus Merci retrievers for thrombectomy revascularisation of large vessel occlusions in acute ischaemic stroke (TREVO 2): a randomised trial. Lancet. 2012;380:1231–1240. doi: 10.1016/S0140-6736(12)61299-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Nolte CH, Doepp F, Schreiber SJ, Gerischer LM, Audebert HJ. Quantification of target population for ultrasound enhanced thrombolysis in acute ischemic stroke. Journal of neuroimaging. 2013;23:79–81. doi: 10.1111/j.1552-6569.2011.00632.x. [DOI] [PubMed] [Google Scholar]
  170. Nonaka Y, Koumura A, Hyakkoku K, Shimazawa M, Yoshimura S, Iwama T, Hara H. Combination treatment with normobaric hyperoxia and cilostazol protects mice against focal cerebral ischemia-induced neuronal damage better than each treatment alone. The Journal of pharmacology and experimental therapeutics. 2009;330:13–22. doi: 10.1124/jpet.109.151548. [DOI] [PubMed] [Google Scholar]
  171. 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. Neuroscience letters. 2008;441:224–228. doi: 10.1016/j.neulet.2008.06.033. [DOI] [PubMed] [Google Scholar]
  172. Ohsawa I, Ishikawa M, Takahashi K, Watanabe M, Nishimaki K, Yamagata K, Katsura K, Katayama Y, Asoh S, Ohta S. Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nature medicine. 2007;13:688–694. doi: 10.1038/nm1577. [DOI] [PubMed] [Google Scholar]
  173. Ono H, Nishijima Y, Adachi N, Sakamoto M, Kudo Y, Kaneko K, Nakao A, Imaoka T. A basic study on molecular hydrogen (H2) inhalation in acute cerebral ischemia patients for safety check with physiological parameters and measurement of blood H2 level. Medical gas research. 2012;2:21. doi: 10.1186/2045-9912-2-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Oron A, Oron U, Chen J, Eilam A, Zhang C, Sadeh M, Lampl Y, Streeter J, DeTaboada L, Chopp M. Low-level laser therapy applied transcranially to rats after induction of stroke significantly reduces long-term neurological deficits. Stroke. 2006;37:2620–2624. doi: 10.1161/01.STR.0000242775.14642.b8. [DOI] [PubMed] [Google Scholar]
  175. Ovesen C, Brizzi M, Pott FC, Thorsen-Meyer HC, Karlsson T, Ersson A, Christensen H, Norrlin A, Meden P, Krieger DW, Petersson J. Feasibility of endovascular and surface cooling strategies in acute stroke. Acta neurologica Scandinavica. 2012 doi: 10.1111/ane.12059. [DOI] [PubMed] [Google Scholar]
  176. Padma MV, Bhasin A, Bhatia R, Garg A, Singh MB, Tripathi M, Prasad K. Normobaric oxygen therapy in acute ischemic stroke: A pilot study in Indian patients. Annals of Indian Academy of Neurology. 2010;13:284–288. doi: 10.4103/0972-2327.74203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Pagel PS, Krolikowski JG, Shim YH, Venkatapuram S, Kersten JR, Weihrauch D, Warltier DC, Pratt PF., Jr Noble gases without anesthetic properties protect myocardium against infarction by activating prosurvival signaling kinases and inhibiting mitochondrial permeability transition in vivo. Anesthesia and analgesia. 2007;105:562–569. doi: 10.1213/01.ane.0000278083.31991.36. [DOI] [PubMed] [Google Scholar]
  178. Pan Y, Zhang H, Acharya AB, Cruz-Flores S, Panneton WM. The effect of heliox treatment in a rat model of focal transient cerebral ischemia. Neuroscience letters. 2011;497:144–147. doi: 10.1016/j.neulet.2011.04.048. [DOI] [PubMed] [Google Scholar]
  179. Pan Y, Zhang H, VanDeripe DR, Cruz-Flores S, Panneton WM. Heliox and oxygen reduce infarct volume in a rat model of focal ischemia. Experimental neurology. 2007;205:587–590. doi: 10.1016/j.expneurol.2007.03.023. [DOI] [PubMed] [Google Scholar]
  180. Pang GS, Wang J, Wang Z, Goh C, Lee CG. The G allele of SNP E1/A118G at the mu-opioid receptor gene locus shows genomic evidence of recent positive selection. Pharmacogenomics. 2009;10:1101–1109. doi: 10.2217/pgs.09.63. [DOI] [PubMed] [Google Scholar]
  181. Park HK, Chu K, Jung KH, Lee ST, Bahn JJ, Kim M, Lee SK, Roh JK. Autophagy is involved in the ischemic p econditioning. Brain research. 2009;1212:71–78. doi: 10.1016/j.neulet.2008.12.019. [DOI] [PubMed] [Google Scholar]
  182. Peng Z, Ren P, Kang Z, Du J, Lian Q, Liu Y, Zhang JH, Sun X. Up-regulated HIF-1alpha is involved in the hypoxic tolerance induced by hyperbaric oxygen preconditioning. Brain research. 2008;1212:71–78. doi: 10.1016/j.brainres.2008.03.027. [DOI] [PubMed] [Google Scholar]
  183. Perez-Pinzon MA, Stetler RA, Fiskum G. Novel mitochondrial targets for neuroprotection. Journal of cerebral blood flow and metabolism. 2012;32:1362–1376. doi: 10.1038/jcbfm.2012.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Preston E, Webster J. A two-hour window for hypothermic modulation of early events that impact delayed opening of the rat blood-brain barrier after ischemia. Acta neuropathologica. 2004;108:406–412. doi: 10.1007/s00401-004-0905-4. [DOI] [PubMed] [Google Scholar]
  185. Puyal J, Ginet V, Clarke PG. Multiple interacting cell death mechanisms in the mediation of excitotoxicity and ischemic brain damage: a challenge for neuroprotection. Progress in neurobiology. 2013;105:24–48. doi: 10.1016/j.pneurobio.2013.03.002. [DOI] [PubMed] [Google Scholar]
  186. Qi ZF, Luo YM, Liu XR, Wang RL, Zhao HP, Yan F, Song ZJ, Luo M, Ji XM. AKT/GSK3beta-Dependent Autophagy Contributes to the Neuroprotection of Limb Remote Ischemic Postconditioning in the Transient Cerebral Ischemic Rat Model. CNS neuroscience & therapeutics. 2012;18:965–973. doi: 10.1111/cns.12016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Ren C, Gao M, Dornbos D, 3rd, Ding Y, Zeng X, Luo Y, Ji X. Remote ischemic post-conditioning reduced brain damage in experimental ischemia/reperfusion injury. Neurological research. 2011;33:514–519. doi: 10.1179/016164111X13007856084241. [DOI] [PubMed] [Google Scholar]
  188. Rockswold SB, Rockswold GL, Zaun DA, Zhang X, Cerra CE, Bergman TA, Liu J. A prospective, randomized clinical trial to compare the effect of hyperbaric to normobaric hyperoxia on cerebral metabolism, intracranial pressure, and oxygen toxicity in severe traumatic brain injury. Journal of neurosurgery. 2010;112:1080–1094. doi: 10.3171/2009.7.JNS09363. [DOI] [PubMed] [Google Scholar]
  189. Ronning OM, Guldvog B. Should stroke victims routinely receive supplemental oxygen? A quasi-randomized controlled trial. Stroke. 1999;30:2033–2037. doi: 10.1161/01.str.30.10.2033. [DOI] [PubMed] [Google Scholar]
  190. Rosenberg GA. Neurological diseases in relation to the blood-brain barrier. Journal of cerebral blood flow and metabolism. 2012;32:1139–1151. doi: 10.1038/jcbfm.2011.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Rusyniak DE, Kirk MA, May JD, Kao LW, Brizendine EJ, Welch JL, Cordell WH, Alonso RJ. Hyperbaric oxygen therapy in acute ischemic stroke: results of the Hyperbaric Oxygen in Acute Ischemic Stroke Trial Pilot Study. Stroke. 2003;34:571–574. doi: 10.1161/01.str.0000050644.48393.d0. [DOI] [PubMed] [Google Scholar]
  192. Ryang YM, Fahlenkamp AV, Rossaint R, Wesp D, Loetscher PD, Beyer C, Coburn M. Neuroprotective effects of argon in an in vivo model of transient middle cerebral artery occlusion in rats. Critical care medicine. 2011;39:1448–1453. doi: 10.1097/CCM.0b013e31821209be. [DOI] [PubMed] [Google Scholar]
  193. Saver JL, Jahan R, Levy EI, Jovin TG, Baxter B, Nogueira RG, Clark W, Budzik R, Zaidat OO. Solitaire flow restoration device versus the Merci Retriever in patients with acute ischaemic stroke (SWIFT): a randomised, parallel-group, non-inferiority trial. Lancet. 2012;380:1241–1249. doi: 10.1016/S0140-6736(12)61384-1. [DOI] [PubMed] [Google Scholar]
  194. Schaechter JD, Connell BD, Stason WB, Kaptchuk TJ, Krebs DE, Macklin EA, Schnyer RN, Stein J, Scarborough DM, Parker SW, McGibbon CA, Wayne PM. Correlated change in upper limb function and motor cortex activation after verum and sham acupuncture in patients with chronic stroke. J Altern Complement Med. 2007;13:527–532. doi: 10.1089/acm.2007.6316. [DOI] [PubMed] [Google Scholar]
  195. Schaller B, Graf R. Hypothermia and stroke: the pathophysiological background. Pathophysiology. 2003;10:7–35. doi: 10.1016/j.pathophys.2003.09.001. [DOI] [PubMed] [Google Scholar]
  196. Schaller B, Graf R, Jacobs AH. Ischaemic tolerance: a window to endogenous neuroprotection? Lancet. 2003;362:1007–1008. doi: 10.1016/S0140-6736(03)14446-7. [DOI] [PubMed] [Google Scholar]
  197. Schwarzenberg H, Muller-Hulsbeck S, Brossman J, Gluer CC, Bruhn HD, Heller M. Hyperthermic fibrinolysis with rt-PA: in vitro results. Cardiovascular and interventional radiology. 1998;21:142–145. doi: 10.1007/s002709900231. [DOI] [PubMed] [Google Scholar]
  198. Sena ES, Jeffreys AL, Cox SF, Sastra SA, Churilov L, Rewell S, Batchelor PE, Vander Worp HB, Macleod MR, Howells DW. The benefit of hypothermia in experimental ischemic stroke is not affected by pethidine. International journal of stroke. 2012;8:180–185. doi: 10.1111/j.1747-4949.2012.00834.x. [DOI] [PubMed] [Google Scholar]
  199. Sheng R, Liu XQ, Zhang LS, Gao B, Han R, Wu YQ, Zhang XY, Qin ZH. Autophagy regulates endoplasmic reticulum stress in ischemic preconditioning. Autophagy. 2012a;8:310–325. doi: 10.4161/auto.18673. [DOI] [PubMed] [Google Scholar]
  200. Sheng R, Zhang LS, Han R, Liu XQ, Gao B, Qin ZH. Autophagy activation is associated with neuroprotection in a rat model of focal cerebral ischemic preconditioning. Autophagy. 2010;6:482–494. doi: 10.4161/auto.6.4.11737. [DOI] [PubMed] [Google Scholar]
  201. Sheng SP, Lei B, James ML, Lascola CD, Venkatraman TN, Jung JY, Maze M, Franks NP, Pearlstein RD, Sheng H, Warner DS. Xenon neuroprotection in experimental stroke: interactions with hypothermia and intracerebral hemorrhage. Anesthesiology. 2012b;117:1262–1275. doi: 10.1097/ALN.0b013e3182746b81. [DOI] [PubMed] [Google Scholar]
  202. Shimizu M, Saxena P, Konstantinov IE, Cherepanov V, Cheung MM, Wearden P, Zhangdong H, Schmidt M, Downey GP, Redington AN. Remote ischemic preconditioning decreases adhesion and selectively modifies functional responses of human neutrophils. The Journal of surgical research. 2010;158:155–161. doi: 10.1016/j.jss.2008.08.010. [DOI] [PubMed] [Google Scholar]
  203. Shimohata T, Zhao H, Sung JH, Sun G, Mochly-Rosen D, Steinberg GK. Suppression of deltaPKC activation after focal cerebral ischemia contributes to the protective effect of hypothermia. Journal of cerebral blood flow and metabolism. 2007;27:1463–1475. doi: 10.1038/sj.jcbfm.9600450. [DOI] [PubMed] [Google Scholar]
  204. Shin HK, Dunn AK, Jones PB, Boas DA, Lo EH, Moskowitz MA, Ayata C. Normobaric hyperoxia improves cerebral blood flow and oxygenation, and inhibits peri-infarct depolarizations in experimental focal ischaemia. Brain. 2007;130:1631–1642. doi: 10.1093/brain/awm071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Si QM, Wu GC, Cao XD. Effects of electroacupuncture on acute cerebral infarction. Acupuncture & electro-therapeutics research. 1998;23:117–124. doi: 10.3727/036012998816356562. [DOI] [PubMed] [Google Scholar]
  206. Sims NR, Muyderman H. Mitochondria, oxidative metabolism and cell death in stroke. Biochimica et biophysica acta. 2010;1802:80–91. doi: 10.1016/j.bbadis.2009.09.003. [DOI] [PubMed] [Google Scholar]
  207. Singhal AB, Benner T, Roccatagliata L, Koroshetz WJ, Schaefer PW, Lo EH, Buonanno FS, Gonzalez RG, Sorensen AG. A pilot study of normobaric oxygen therapy in acute ischemic stroke. Stroke. 2005a;36:797–802. doi: 10.1161/01.STR.0000158914.66827.2e. [DOI] [PubMed] [Google Scholar]
  208. Singhal AB, Dijkhuizen RM, Rosen BR, Lo EH. Normobaric hyperoxia reduces MRI diffusion abnormalities and infarct size in experimental stroke. Neurology. 2002a;58:945–952. doi: 10.1212/wnl.58.6.945. [DOI] [PubMed] [Google Scholar]
  209. Singhal AB, Lo EH, Dalkara T, Moskowitz MA. Advances in stroke neuroprotection: hyperoxia and beyond. Neuroimaging clinics of North America. 2005b;15:697–720. xii–xiii. doi: 10.1016/j.nic.2005.08.014. [DOI] [PubMed] [Google Scholar]
  210. Singhal AB, Ratai E, Benner T, Vangel M, Lee V, Koroshetz WJ, Schaefer PW, Sorensen AG, Gonzalez RG. Magnetic resonance spectroscopy study of oxygen therapy in ischemic stroke. Stroke. 2007;38:2851–2854. doi: 10.1161/STROKEAHA.107.487280. [DOI] [PubMed] [Google Scholar]
  211. Singhal AB, Wang X, Sumii T, Mori T, Lo EH. Effects of normobaric hyperoxia in a rat model of focal cerebral ischemia-reperfusion. Journal of cerebral blood flow and metabolism. 2002b;22:861–868. doi: 10.1097/00004647-200207000-00011. [DOI] [PubMed] [Google Scholar]
  212. Soejima Y, Hu Q, Krafft PR, Fujii M, Tang J, Zhang JH. Hyperbaric oxygen preconditioning attenuates hyperglycemia-enhanced hemorrhagic transformation by inhibiting matrix metalloproteinases in focal cerebral ischemia in rats. Experimental neurology. 2013;247:737–743. doi: 10.1016/j.expneurol.2013.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Soejima Y, Ostrowski RP, Manaenko A, Fujii M, Tang J, Zhang JH. Hyperbaric oxygen preconditioning attenuates hyperglycemia enhanced hemorrhagic transformation after transient MCAO in rats. Medical gas research. 2012;2:9. doi: 10.1186/2045-9912-2-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Staat P, Rioufol G, Piot C, Cottin Y, Cung TT, L’Huillier I, Aupetit JF, Bonnefoy E, Finet G, Andre-Fouet X, Ovize M. Postconditioning the human heart. Circulation. 2005;112:2143–2148. doi: 10.1161/CIRCULATIONAHA.105.558122. [DOI] [PubMed] [Google Scholar]
  215. (STAIR), S. T. A. I. R. Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke. 1999;30:2752–2758. doi: 10.1161/01.str.30.12.2752. [DOI] [PubMed] [Google Scholar]
  216. Su DS, Wang ZH, Zheng YJ, Zhao YH, Wang XR. Dose-dependent neuroprotection of delta opioid peptide [D-Ala2, D-Leu5] enkephalin in neuronal death and retarded behavior induced by forebrain ischemia in rats. Neuroscience letters. 2007;423:113–117. doi: 10.1016/j.neulet.2007.06.044. [DOI] [PubMed] [Google Scholar]
  217. Sun L, Strelow H, Mies G, Veltkamp R. Oxygen therapy improves energy metabolism in focal cerebral ischemia. Brain research. 2011;1415:103–108. doi: 10.1016/j.brainres.2011.07.064. [DOI] [PubMed] [Google Scholar]
  218. Sung JH, Chao DM, Xia Y. Neuronal responses to hypoxia. In: Wang Q, Ying WH, editors. New Frontiers in Neurological Research. Kerala: Research Signpost; 2008. [Google Scholar]
  219. Sutherland BA, Harrison JC, Nair SM, Sammut IA. Inhalation gases or gaseous mediators as neuroprotectants for cerebral ischaemia. Current drug targets. 2013;14:56–73. doi: 10.2174/138945013804806433. [DOI] [PubMed] [Google Scholar]
  220. Szczudlik A, Turaj W, Slowik A, Strojny J. Microalbuminuria and hyperthermia independently predict long-term mortality in acute ischemic stroke patients. Acta neurologica Scandinavica. 2003;107:96–101. doi: 10.1034/j.1600-0404.2003.01363.x. [DOI] [PubMed] [Google Scholar]
  221. Tachibana K, Hansmann DR, Anderson JR. Ultrasound catheter for providing a therapeutic effect to a vessel of the body. 6001069. US. 1999
  222. Tam WK, Tong KY, Meng F, Gao S. A minimal set of electrodes for motor imagery BCI to control an assistive device in chronic stroke subjects: a multi-session study. IEEE transactions on neural systems and rehabilitation engineering. 2011;19:617–627. doi: 10.1109/TNSRE.2011.2168542. [DOI] [PubMed] [Google Scholar]
  223. Thompson JW, Narayanan SV, Perez-Pinzon MA. Redox signaling pathways involved in neuronal ischemic preconditioning. Current neuropharmacology. 2012;10:354–369. doi: 10.2174/157015912804143577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Thomas LH, Cross S, Barrett J, French B, Leathley M, Sutton CJ, Watkins C. Treatment of urinary incontinence after stroke in adults. Cochrane Database Syst Rev. 2008;23:CD004462. doi: 10.1002/14651858.CD004462.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Tian X, Guo J, Zhu M, Li M, Wu G, Xia Y. delta-Opioid receptor activation rescues the functional TrkB receptor and protects the brain from ischemia-reperfusion injury in the rat. PloS one. 2013a;8:e69252. doi: 10.1371/journal.pone.0069252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Tian X, Hua F, Sandhu HK, Chao D, Balboni G, Salvadori S, He X, Xia Y. Effect of delta-opioid receptor activation on BDNF-TrkB vs. TNF-alpha in the mouse cortex exposed to prolonged hypoxia. International journal of molecular sciences. 2013b;14:15959–15976. doi: 10.3390/ijms140815959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Tian XS, Zhou F, Yang R, Xia Y, Wu GC, Guo JC. Effects of intracerebroventricular injection of delta-opioid receptor agonist TAN-67 or antagonist naltrindole on acute cerebral ischemia in rat. Acta physiologica Sinica. 2008a;60:475–484. [PubMed] [Google Scholar]
  228. Tian XS, Zhou F, Yang R, Xia Y, Wu GC, Guo JC. Electro-acupuncture protects the brain against ischemic injury via up-regulation of delta-opioid receptors. J Chin Integr Med. 2008b;6:632–638. doi: 10.3736/jcim20080617. [DOI] [PubMed] [Google Scholar]
  229. Tsai JY, Pan W, Lemaire SA, Pisklak P, Lee VV, Bracey AW, Elayda MA, Preventza O, Price MD, Collard CD, Coselli JS. Moderate hypothermia during aortic arch surgery is associated with reduced risk of early mortality. The Journal of thoracic and cardiovascular surgery. 2013;146:662–667. doi: 10.1016/j.jtcvs.2013.03.004. [DOI] [PubMed] [Google Scholar]
  230. Uozumi Y, Nawashiro H, Sato S, Kawauchi S, Shima K, Kikuchi M. Targeted increase in cerebral blood flow by transcranial near-infrared laser irradiation. Lasers in surgery and medicine. 2010;42:566–576. doi: 10.1002/lsm.20938. [DOI] [PubMed] [Google Scholar]
  231. Van der Worp HB, Sena ES, Donnan GA, Howells DW, Macleod MR. Hypothermia in animal models of acute ischaemic stroke: a systematic review and meta-analysis. Brain. 2007;130:3063–3074. doi: 10.1093/brain/awm083. [DOI] [PubMed] [Google Scholar]
  232. Varon J, Marik PE, Einav S. Therapeutic hypothermia: a state-of-the-art emergency medicine perspective. The American journal of emergency medicine. 2012;30:800–810. doi: 10.1016/j.ajem.2011.03.007. [DOI] [PubMed] [Google Scholar]
  233. Veltkamp R, Siebing DA, Heiland S, Schoenffeldt-Varas P, Veltkamp C, Schwaninger M, Schwab S. Hyperbaric oxygen induces rapid protection against focal cerebral ischemia. Brain research. 2005;1037:134–138. doi: 10.1016/j.brainres.2005.01.006. [DOI] [PubMed] [Google Scholar]
  234. Wacker BK, Freie AB, Perfater JL, Gidday JM. Junctional protein regulation by sphingosine kinase 2 contributes to blood-brain barrier protection in hypoxic preconditioning-induced cerebral ischemic tolerance. Journal of cerebral blood flow and metabolism. 2012;32:1014–1023. doi: 10.1038/jcbfm.2012.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Wada K, Ito M, Miyazawa T, Katoh H, Nawashiro H, Shima K, Chigasaki H. Repeated hyperbaric oxygen induces ischemic tolerance in gerbil hippocampus. Brain research. 1996;740:15–20. doi: 10.1016/s0006-8993(96)00831-1. [DOI] [PubMed] [Google Scholar]
  236. Wagner KR, Beiler S, Beiler C, Kirkman J, Casey K, Robinson T, Larnard D, de Courten-Myers GM, Linke MJ, Zuccarello M. Delayed profound local brain hypothermia markedly reduces interleukin-1beta gene expression and vasogenic edema development in a porcine model of intracerebral hemorrhage. Acta neurochirurgica. Supplement. 2006;96:177–182. doi: 10.1007/3-211-30714-1_39. [DOI] [PubMed] [Google Scholar]
  237. Wagner S, Nagel S, Kluge B, Schwab S, Heiland S, Koziol J, Gardner H, Hacke W. Topographically graded postischemic presence of metalloproteinases is inhibited by hypothermia. Brain research. 2003;984:63–75. doi: 10.1016/s0006-8993(03)03088-9. [DOI] [PubMed] [Google Scholar]
  238. Wang H, Olivero W, Lanzino G, Elkins W, Rose J, Honings D, Rodde M, Burnham J, Wang D. Rapid and selective cerebral hypothermia achieved using a cooling helmet. Journal of neurosurgery. 2004;100:272–277. doi: 10.3171/jns.2004.100.2.0272. [DOI] [PubMed] [Google Scholar]
  239. Wang JY, Shen J, Gao Q, Ye ZG, Yang SY, Liang HW, Bruce IC, Luo BY, Xia Q. Ischemic postconditioning protects against global cerebral ischemia/reperfusion-induced injury in rats. Stroke. 2008;39:983–990. doi: 10.1161/STROKEAHA.107.499079. [DOI] [PubMed] [Google Scholar]
  240. Wang SM, Lin EC, Maranets I, Kain ZN. The impact of asynchronous electroacupuncture stimulation duration on cold thermal pain threshold. Anesthesia and analgesia. 2009;109:932–935. doi: 10.1213/ane.0b013e3181ad9292. [DOI] [PubMed] [Google Scholar]
  241. Wei D, Ren C, Chen X, Zhao H. The chronic protective effects of limb remote preconditioning and the underlying mechanisms involved in inflammatory factors in rat stroke. PloS one. 2012;7:e30892. doi: 10.1371/journal.pone.0030892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Wei M, Xin P, Li S, Tao J, Li Y, Li J, Liu M, Zhu W, Redington AN. Repeated remote ischemic postconditioning protects against adverse left ventricular remodeling and improves survival in a rat model of myocardial infarction. Circulation research. 2011;108:1220–1225. doi: 10.1161/CIRCRESAHA.110.236190. [DOI] [PubMed] [Google Scholar]
  243. Wen GQ, He XZ, Lu Y, Xia Y. Effect of acupuncture on neurotransmitters/modulators. In: Xia, et al., editors. Acupuncture Therapy for Neurological Diseases: A Neurobiological View. Springer-Tsinghua Press; Heidelberg, London, New York, Beijing: 2010a. pp. 120–142. [Google Scholar]
  244. Wen GQ, Yang YL, Lu Y, Xia Y. Acupuncture-induced activation of endogenous opioid system. In: Xia, et al., editors. Acupuncture Therapy for Neurological Diseases: A Neurobiological View. Springer-Tsinghua Press; Heidelberg, London, New York, Beijing: 2010b. pp. 104–119. [Google Scholar]
  245. Wilkie KM, Shiels JE, Bulley C, Salisbury LG. “Functional electrical stimulation (FES) impacted on important aspects of my life”: a qualitative exploration of chronic stroke patients’ and carers’ perceptions of FES in the management of dropped foot. Physiotherapy theory and practice. 2012;28:1–9. doi: 10.3109/09593985.2011.563775. [DOI] [PubMed] [Google Scholar]
  246. Wong AM, Su TY, Tang FT, Cheng PT, Liaw MY. Clinical trial of electrical acupuncture on hemiplegic stroke patients. American journal of physical medicine & rehabilitation/Association of Academic Physiatrists. 1999;78:117–122. doi: 10.1097/00002060-199903000-00006. [DOI] [PubMed] [Google Scholar]
  247. World Health Organization. Acupuncture: review and analysis of reports on controlled clinical trials. 2003 (NLM classification: WB 369) [Google Scholar]
  248. Wu O, Lu J, Mandeville JB, Murata Y, Egi Y, Dai G, Marota JJ, Diwan I, Dijkhuizen RM, Kwong KK, Lo EH, Singhal AB. Dynamic functional cerebral blood volume responses to normobaric hyperoxia in acute ischemic stroke. Journal of cerebral blood flow and metabolism. 2012;32:1800–1809. doi: 10.1038/jcbfm.2012.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Wu TC, Grotta JC. Hypothermia for acute ischaemic stroke. Lancet neurology. 2013;12:275–284. doi: 10.1016/S1474-4422(13)70013-9. [DOI] [PubMed] [Google Scholar]
  250. Xia Y, Cao XD, Wu GC, Cheng JS. Acupuncture Therapy for Neurological Diseases: A Neurobiological View. Springer-Tsinghua Press; Beijing, Heidelberg, London, New York: 2010. pp. 1–480. [Google Scholar]
  251. Xia Y, Ding GH, Wu GC. Current Research in Acupuncture. Springer; New York, Heidelberg, Dordrecht, London: 2012. pp. 1–752. [Google Scholar]
  252. Xing B, Chen H, Zhang M, Zhao D, Jiang R, Liu X, Zhang S. Ischemic postconditioning protects brain and reduces inflammation in a rat model of focal cerebral ischemia/reperfusion. Journal of neurochemistry. 2008;105:1737–1745. doi: 10.1111/j.1471-4159.2008.05276.x. [DOI] [PubMed] [Google Scholar]
  253. Xiong LZ, Yang J, Wang Q, Lu ZH. Involvement of δ-and μ-opioid receptors in the delayed cerebral ischemic tolerance induced by repeated electroacupuncture preconditioning in rats. Chinese Medical Journal. 2007;120:394–399. [PubMed] [Google Scholar]
  254. Xu MS, Ge LB, Zhao D. Electro-acupuncture Regulation of Central Monoamine Neurotransmitters in Ischaemia-Reperfusion. In: Xia, et al., editors. Current Research in Acupuncture. Springer; New York, Heidelberg, Dordrecht, London: 2012. pp. 401–430. [Google Scholar]
  255. Yan W, Zhang H, Bai X, Lu Y, Dong H, Xiong L. Autophagy activation is involved in neuroprotection induced by hyperbaric oxygen preconditioning against focal cerebral ischemia in rats. Brain research. 2011;1402:109–121. doi: 10.1016/j.brainres.2011.05.049. [DOI] [PubMed] [Google Scholar]
  256. Yang Y, Xia X, Zhang Y, Wang Q, Li L, Luo G, Xia Y. delta-Opioid receptor activation attenuates oxidative injury in the ischemic rat brain. BMC biology. 2009;7:55. doi: 10.1186/1741-7007-7-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Yang Y, Rosenberg GA. MMP-mediated disruption of claudin-5 in the blood-brain barrier of rat brain after cerebral ischemia. Methods Mol Biol. 2011;762:333–345. doi: 10.1007/978-1-61779-185-7_24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Yang ZJ, Xie Y, Bosco GM, Chen C, Camporesi EM. Hyperbaric oxygenation alleviates MCAO-induced brain injury and reduces hydroxyl radical formation and glutamate release. European journal of applied physiology. 2010;108:513–522. doi: 10.1007/s00421-009-1229-9. [DOI] [PubMed] [Google Scholar]
  259. Yenari MA, Han HS. Influence of hypothermia on post-ischemic inflammation: role of nuclear factor kappa B (NFkappaB) Neurochemistry international. 2006;49:164–169. doi: 10.1016/j.neuint.2006.03.016. [DOI] [PubMed] [Google Scholar]
  260. Yenari MA, Han HS. Neuroprotective mechanisms of hypothermia in brain ischaemia. Nature reviews. Neuroscience. 2012;13:267–278. doi: 10.1038/nrn3174. [DOI] [PubMed] [Google Scholar]
  261. Yenari MA, Iwayama S, Cheng D, Sun GH, Fujimura M, Morita-Fujimura Y, Chan PH, Steinberg GK. Mild hypothermia attenuates cytochrome c release but does not alter Bcl-2 expression or caspase activation after experimental stroke. Journal of cerebral blood flow and metabolism. 2002;22:29–38. doi: 10.1097/00004647-200201000-00004. [DOI] [PubMed] [Google Scholar]
  262. Yenari MA, Palmer JT, Bracci PM, Steinberg GK. Thrombolysis with tissue plasminogen activator (tPA) is temperature dependent. Thrombosis research. 1995;77:475–481. doi: 10.1016/0049-3848(95)93883-2. [DOI] [PubMed] [Google Scholar]
  263. 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 research. 2010;1352:248–254. doi: 10.1016/j.brainres.2010.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Zeiner A, Holzer M, Sterz F, Behringer W, Schorkhuber W, Mullner M, Frass M, Siostrzonek P, Ratheiser K, Kaff A, Laggner AN. Mild resuscitative hypothermia to improve neurological outcome after cardiac arrest. A clinical feasibility trial. Hypothermia After Cardiac Arrest (HACA) Study Group. Stroke. 2000;31:86–94. doi: 10.1161/01.str.31.1.86. [DOI] [PubMed] [Google Scholar]
  265. Zeng Y, Xie K, Dong H, Zhang H, Wang F, Li Y, Xiong L. Hyperbaric oxygen preconditioning protects cortical neurons against oxygen-glucose deprivation injury: role of peroxisome proliferator-activated receptor-gamma. Brain research. 2012;1452:140–150. doi: 10.1016/j.brainres.2012.02.063. [DOI] [PubMed] [Google Scholar]
  266. Zhang J, Gibney GT, Zhao P, Xia Y. Neuroprotective role of delta-opioid receptors in cortical neurons. American journal of physiology. 2002;282:C1225–1234. doi: 10.1152/ajpcell.00226.2001. [DOI] [PubMed] [Google Scholar]
  267. Zhang J, Haddad GG, Xia Y. delta-, but not mu- and kappa-, opioid receptor activation protects neocortical neurons from glutamate-induced excitotoxic injury. Brain research. 2000;885:143–153. doi: 10.1016/s0006-8993(00)02906-1. [DOI] [PubMed] [Google Scholar]
  268. Zhang J, Qian H, Zhao P, Hong SS, Xia Y. Rapid hypoxia preconditioning protects cortical neurons from glutamate toxicity through delta-opioid receptor. Stroke. 2006;37:1094–1099. doi: 10.1161/01.STR.0000206444.29930.18. [DOI] [PubMed] [Google Scholar]
  269. Zhang J, Menniti-Ippolito F, Gao X, Firenzuoli F, Zhang B, Massari M, Shang H, Huang Y, Ferrelli R, Hu L, Fauci A, Guerra R, Raschetti R. Complex Traditional Chinese Medicine for Poststroke Motor Dysfunction: A Systematic Review. Stroke. 2009;40:2797–2804. doi: 10.1161/STROKEAHA.109.555227. [DOI] [PubMed] [Google Scholar]
  270. Zhang JH, Xia Y, Haddad GG. Activation of δ-opioid receptors protects cortical neurons from glutamate excitotoxic injury. Soc Neurosci. 1999;28:736. [Google Scholar]
  271. Zhang RL, Chopp M, Chen H, Garcia JH, Zhang ZG. Postischemic (1 hour) hypothermia significantly reduces ischemic cell damage in rats subjected to 2 hours of middle cerebral artery occlusion. Stroke. 1993;24:1235–1240. doi: 10.1161/01.str.24.8.1235. [DOI] [PubMed] [Google Scholar]
  272. Zhang Y, Park TS, Gidday JM. Hypoxic preconditioning protects human brain endothelium from ischemic apoptosis by Akt-dependent survivin activation. American journal of physiology. Heart and circulatory physiology. 2007;292:H2573–2581. doi: 10.1152/ajpheart.01098.2006. [DOI] [PubMed] [Google Scholar]
  273. Zhao H. Ischemic postconditioning as a novel avenue to protect against brain injury after stroke. Journal of cerebral blood flow and metabolism. 2009;29:873–885. doi: 10.1038/jcbfm.2009.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Zhao H, Sapolsky RM, Steinberg GK. Interrupting reperfusion as a stroke therapy: ischemic postconditioning reduces infarct size after focal ischemia in rats. Journal of cerebral blood flow and metabolism. 2006;26:1114–1121. doi: 10.1038/sj.jcbfm.9600348. [DOI] [PubMed] [Google Scholar]
  275. Zhao H, Steinberg GK, Sapolsky RM. General versus specific actions of mild-moderate hypothermia in attenuating cerebral ischemic damage. Journal of cerebral blood flow and metabolism. 2007a;27:1879–1894. doi: 10.1038/sj.jcbfm.9600540. [DOI] [PubMed] [Google Scholar]
  276. Zhao H, Wang JQ, Shimohata T, Sun G, Yenari MA, Sapolsky RM, Steinberg GK. Conditions of protection by hypothermia and effects on apoptotic pathways in a rat model of permanent middle cerebral artery occlusion. Journal of neurosurgery. 2007b;107:636–641. doi: 10.3171/JNS-07/09/0636. [DOI] [PubMed] [Google Scholar]
  277. Zhao L, Liu X, Liang J, Han S, Wang Y, Yin Y, Luo Y, Li J. Phosphorylation of p38 MAPK mediates hypoxic preconditioning-induced neuroprotection against cerebral ischemic injury via mitochondria translocation of Bcl-xL in mice. Brain research. 2013;1503:78–88. doi: 10.1016/j.brainres.2013.01.051. [DOI] [PubMed] [Google Scholar]
  278. Zhao L, Nowak TS., Jr CBF changes associated with focal ischemic preconditioning in the spontaneously hypertensive rat. Journal of cerebral blood flow and metabolism. 2006;26:1128–1140. doi: 10.1038/sj.jcbfm.9600269. [DOI] [PubMed] [Google Scholar]
  279. Zhao P, Guo JC, Hong SS, Bazzy-Assad A, Cheng JS, Xia Y. Electro-acupuncture and brain protection from cerebral ischemia: The role of δ-opioid receptor. Society for Neuroscience Abstract. 2002;28:736. [Google Scholar]
  280. Zhao ZQ, Corvera JS, Halkos ME, Kerendi F, Wang NP, Guyton RA, Vinten-Johansen J. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. American journal of physiology. Heart and circulatory physiology. 2003;285:H579–588. doi: 10.1152/ajpheart.01064.2002. [DOI] [PubMed] [Google Scholar]
  281. Zhao ZQ, Vinten-Johansen J. Postconditioning: reduction of reperfusion-induced injury. Cardiovascular research. 2006;70:200–211. doi: 10.1016/j.cardiores.2006.01.024. [DOI] [PubMed] [Google Scholar]
  282. Zheng Z, Kim JY, Ma H, Lee JE, Yenari MA. Anti-inflammatory effects of the 70 kDa heat shock protein in experimental stroke. Journal of cerebral blood flow and metabolism. 2008;28:53–63. doi: 10.1038/sj.jcbfm.9600502. [DOI] [PubMed] [Google Scholar]
  283. Zhou C, Tu J, Zhang Q, Lu D, Zhu Y, Zhang W, Yang F, Brann DW, Wang R. Delayed ischemic postconditioning protects hippocampal CA1 neurons by preserving mitochondrial integrity via Akt/GSK3beta signaling. Neurochemistry international. 2011a;59:749–758. doi: 10.1016/j.neuint.2011.08.008. [DOI] [PubMed] [Google Scholar]
  284. Zhou F, Guo J, Cheng J, Wu G, Sun J, Xia Y. Electroacupuncture and Brain Protection against Cerebral Ischemia: Specific Effects of Acupoints. Evidence-based complementary and alternative medicine. 2013a;2013:804397. doi: 10.1155/2013/804397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Zhou F, Guo J, Cheng J, Wu G, Xia Y. Chinese herbal medicine or electro-acupuncture: Which is better for brain protection from cerebral ischemia?. The Proceddings of the 7th meeting of the Consortium for Globalization of Chinese Medicine, Bio-II-104; 2008. p. 149. [Google Scholar]
  286. Zhou F, Guo J, Cheng J, Wu G, Xia Y. Electroacupuncture increased cerebral blood flow and reduced ischemic brain injury: dependence on stimulation intensity and frequency. J Appl Physiol. 2011b;111:1877–1887. doi: 10.1152/japplphysiol.00313.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Zhou F, Guo J, Cheng J, Wu G, Xia Y. Effect of electroacupuncture on rat ischemic brain injury: importance of stimulation duration. Evidence-based complementary and alternative medicine. 2013b;2013:878521. doi: 10.1155/2013/878521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Zhou F, Huang DK, Xia Y. Acupuncture Therapy for Neurological Diseases: A Neurobiological View. Springer-Tsinghua Press; Heidelberg, London, New York, Beijing: 2010. Neuroanatomic basis of acupuncture points; pp. 32–80. [Google Scholar]
  289. Zhuang L, Yang T, Zhao H, Fidalgo AR, Vizcaychipi MP, Sanders RD, Yu B, Takata M, Johnson MR, Ma D. The protective profile of argon, helium, and xenon in a model of neonatal asphyxia in rats. Critical care medicine. 2012;40:1724–1730. doi: 10.1097/CCM.0b013e3182452164. [DOI] [PubMed] [Google Scholar]
  290. Zivin JA, Albers GW, Bornstein N, Chippendale T, Dahlof B, Devlin T, Fisher M, Hacke W, Holt W, Ilic S, Kasner S, Lew R, Nash M, Perez J, Rymer M, Schellinger P, Schneider D, Schwab S, Veltkamp R, Walker M, Streeter J. Effectiveness and safety of transcranial laser therapy for acute ischemic stroke. Stroke. 2009;40:1359–1364. doi: 10.1161/STROKEAHA.109.547547. [DOI] [PubMed] [Google Scholar]
  291. Zivin JA, Sehra R, Shoshoo A, Albers GW, Bornstein NM, Dahlof B, Kasner SE, Howard G, Shuaib A, Streeter J, Richieri SP, Hacke W. NeuroThera((R)) Efficacy and Safety Trial - 3 (NEST-3): a double-blind, randomized, sham-controlled, parallel group, multicenter, pivotal study to assess the safety and efficacy of transcranial laser therapy with the NeuroThera((R)) Laser System for the treatment of acute ischemic stroke within 24 h of stroke onset. International journal of stroke. 2012 Sep 27; doi: 10.1111/j.1747-4949.2012.00896.x. Epub ahead of print. [DOI] [PubMed] [Google Scholar]

RESOURCES