• Users Online: 1666
  • Home
  • Print this page
  • Email this page
Home About us Editorial board Ahead of print Current issue Search Archives Submit article Instructions Subscribe Contacts Login 

   Table of Contents      
REVIEW ARTICLE
Year : 2015  |  Volume : 1  |  Issue : 1  |  Page : 14-25

Hypothermia for treatment of stroke


Department of Neurology, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, California, USA

Date of Submission12-Apr-2015
Date of Acceptance08-Aug-2015
Date of Web Publication30-Sep-2015

Correspondence Address:
Midori A Yenari
Neurology 127, VAMC, 4150 Clement St., San Francisco, California - 94121
USA
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/2394-8108.164997

Rights and Permissions
  Abstract 

Stroke is a major cause of neurological disability and death in industrialized nations. Therapeutic hypothermia has been shown to protect the brain from ischemia, stroke, and other acute neurological insults at the laboratory level. It has been shown to improve neurological outcome in certain clinical settings including anoxic brain injury due to cardiac arrest and hypoxic-ischemic neonatal encephalopathy. Hypothermia seems to affect multiple aspects of brain physiology and it is likely that multiple mechanisms underlie its protective effect. Understanding the events that occur in the ischemic brain during hypothermia might help lead to an understanding of how to protect the brain against acute injuries.

Keywords: Apoptosis, blood-brain barrier (BBB) and edema, hypoxic-ischemic injury, inflammation, intracerebral hemorrhage (ICH), multiple mechanisms, neuroprotection, subarachnoid hemorrhage (SAH)


How to cite this article:
Kim JY, Yenari MA. Hypothermia for treatment of stroke. Brain Circ 2015;1:14-25

How to cite this URL:
Kim JY, Yenari MA. Hypothermia for treatment of stroke. Brain Circ [serial online] 2015 [cited 2023 Jun 3];1:14-25. Available from: http://www.braincirculation.org/text.asp?2015/1/1/14/164997


  Introduction Top


Several laboratories have shown over the years that hypothermia protects the brain from various insults such as stroke, hypoxia ischemia, forebrain ischemia, neonatal hypoxia-ischemia, and brain trauma. It is an effective therapy against brain cell death, especially when cooling is initiated within a few hours of injury onset. Therapeutic hypothermia has been extensively studied at the experimental level, and has been correlated to several salutary effects including reduction of metabolic activity, glutamate release, inflammation, production of reactive oxygen species, and mitochondrial cytochrome c release. [1] Such observations have been shown by several laboratories using different models and methods. Recent clinical studies have established that therapeutic cooling improves neurological outcome from cardiac arrest [2],[3] and neonatal hypoxia-ischemia. [4],[5],[6] There are fewer randomized multicenter trials in stroke patients, with a few studies mostly demonstrating its feasibility [7],[8],[9] and safety. [10],[11] However, there have been no studies demonstrating efficacy in human stroke. Clinical stroke studies seem to be hampered by unique challenges such as effective cooling strategies in awake patients and the potential for adverse events in a typically older patient population. Thus, while hypothermia has been shown to be efficacious in multiple laboratory models of stroke, it has not yet been demonstrated to do the same in humans. Further, hypothermia might be used as a model of neuroprotection by which mechanisms of stroke evolution could be better understood and as a means by which therapeutic targets may be identified. This review will cover basic and preclinical studies of hypothermia in experimental stroke and corresponding clinical studies as well as the many underlying mechanisms of hypothermic brain protection.


  Preclinical Studies and Clinical Correlation Top


Optimal target temperature for therapeutic hypothermia

Therapeutic hypothermia is defined as an intentionally induced, controlled reduction from a normal body temperature of 37~38°C to temperatures in the range of 32-35°C (mild hypothermia), 28-32°C (moderate hypothermia), 20~28°C (deep hypothermia), and 5-20°C (profound hypothermia).

In earlier studies, deep to profound hypothermia was frequently applied; however, cooling to such low temperatures led to numerous complications, and was also difficult to achieve and maintain. In the late 1980s, there was a renewed interest in therapeutic cooling when studies showed that mild to moderate hypothermia was equally effective and better tolerated. [12] The scientific literature indicates that lowering brain temperature to the mild to moderate range is similarly protective as deep hypothermia. [13],[14] In a meta-analysis of animal studies, the benefits of hypothermia were inversely related to the temperature attained and hypothermia reduced infarct size by >40% with temperatures of 34°C or below. [15] However, specific studies that directly compared different temperatures failed to show dose dependent neuroprotection comparing temperatures of 27°C to 32°C [16] and 30°C to 33°C. [17] In a rodent study of experimental stroke, cooling to temperatures of 36°C, 35°C, 34°C, 33°C, and 32°C for 4 h was carriedout. Cooling to 35°C failed to improve the outcome while the largest benefit was obtained at 34°C. [18]

Most clinical studies of hypothermia in acute ischemic stroke [8],[9],[19] as well as in postanoxic encephalopathy after cardiac arrest, [2] perinatal hypoxic-ischemic encephalopathy (HIE), [20] and traumatic brain injury [21],[22] have studied cooling to levels of 32~34°C. Unfortunately, discomfort and shivering increased with lower temperatures in awake stroke patients, and cooling to these levels therefore required sedation, mechanical ventilation, and admission to an intensive care unit (ICU). [23] On the other hand, cooling patients with severe head injury to 35°C appeared to be as beneficial as 33°C while causing fewer complications. [24] In addition, temperature reductions to 35°C or 35.5°C via surface cooling have been shown to be safe in stroke patients without sedation but these patients were instead provided drugs such as meperidine to prevent shivering. [25] Using a similar strategy, the Nordic Cooling Stroke Study (NoCSS) was a randomized trial that tested the effect of temperature reduction to 35°C in patients who were awake with surface cooling for 9 h, started within 6 h of ischemic stroke onset but the trial was unfortunately terminated owing to slow recruitment.

A recent international clinical study of over 900 comatose survivors of cardiac arrest cooled patients in two separate groups to 33°C and 36°C. After 3 months of follow-up, both the groups had similar outcomes. [26] The results of this study led some groups to speculate that cooling to 36°C was as beneficial as 33°C. Certainly, cooling to 36°C is more feasible and better tolerated by most patients. However, this study did not include a group where normothermia was maintained. Thus, it is unclear whether cooling to 36°C was beneficial compared to no cooling, or if all cooling was ineffective.

In sum, the current literature suggests that cooling to 32~34°C may be the optimal target temperature. Cooling to 35°C may be safe but its neuroprotective potential is less clear.

Treatment window and duration of hypothermia

Numerous laboratory studies have demonstrated the benefit of timing and duration of hypothermia determining the effects of cooling, with early initiation of cooling before the brain injury to confer significant neuroprotective outcome. Though early initiation is not always feasible in clinical settings, it is still important from a treatment perspective to determine the optimal therapeutic window for hypothermia. In a prior review, it was reported that infarct size reduction was often observed when cooling was begun within 60 min of stroke onset in permanent middle cerebral artery (MCA) occlusion models [27] and within 180 min of stroke onset in temporary MCA occlusion models. [28] In global cerebral ischemia, hypothermia delayed by 6 h after ischemia onset was reported to be advantageous compared to normothermic control groups, with CA1 hippocampal cell loss as a histologic endpoint. [29] A study in hippocampal slices subjected to in vitro ischemia showed hippocampal neuron protection when cooling was delayed as late as 8 h. [30] However, similar delays have not been shown for focal cerebral ischemia.

The optimal duration of therapeutic hypothermia is also not known. Some groups have used brief durations of hypothermia (0.5~5 h), whereas others have used longer periods (12~48 h). In a few studies of focal cerebral ischemia where the duration of intraischemic hypothermia was compared directly, durations of 1-3 h appeared effective, whereas durations between 30 min and 1 h were not effective. [17],[31] In global cerebral ischemia, intraischemic hypothermia (rectal temperature 28~32°C) completely prevented hippocampal cell damage if continued for 4 h or 6 h, whereas 2 h of hypothermia protected less well and 1 h or 30 min did not protect at all. [32] Longer durations may be necessary especially when the initiation of cooling is delayed. Some studies showed that postischemic hypothermia merely delayed the onset of irreversible neuronal injury, unless combined with a second neuroprotectant. [33],[34] However, these latter studies only applied hypothermia for 3 h. Other investigators have shown that prolonged hypothermia initiated 4~6 h after forebrain ischemia for 24 h can provide sustained functional and histological neuroprotection as late as 6 months after ischemia onset. [35] Rodent data indicated that prolonged reduction in temperature induced robust neuroprotection when hypothermia is delayed by several hours, provided cooling is maintained for more than 24 h. [29] Thus, the extent of a neuroprotection also appears to be influenced by the length of the delay and the duration of cooling.

In clinical stroke, hypothermia may be a more effective neuroprotection strategy if applied for a long duration after the ischemic event as most patients do not present until hours after the onset of stroke. [36] Although a long cooling period seems attractive, this may be offset by an increased risk of complications. Furthermore, many studies in animal models were carried out for relatively short time periods, and the cooling duration was somewhat brief. Extensive analysis on the methodological aspects of therapeutic cooling raises questions as to whether the observed effects in the laboratory are durable, and whether studies in young laboratory animals apply to older adult animals with various comorbidities. [37],[38]

Hypothermia in brain ischemia

Global cerebral ischemia

Several experimental studies have demonstrated the neuroprotective effects of mild or moderate hypothermia for cardiac arrest (global ischemia). [12],[28] These studies have shown the durability of this protective effect and have defined a temporal therapeutic window that can be lengthened, provided cooling is prolonged. A global ischemia study in gerbils found that cooling in the range of 30~34°C leads to robust neuroprotection. [35] Mild hypothermia for 12 h enhances neuroprotection of hippocampal CA1 after a 3 min insult, whereas neuroprotection was less pronounced after a 5 min insult, unless hypothermia was maintained for 24 h. [39] Thus, longer cooling may be suited for more severe insults.

The clinical benefit of hypothermia has also been demonstrated in two large-scale clinical studies based on data from multiple medical centers conduced in 2002. [2],[3] These clinical studies showed that mild hypothermia for 12~24 h reduces mortality and improves functional recovery from cardiac attest. [2],[3] Cooled patients had improved neurological outcome 6 months later, compared to those who were normothermic. [3] Since then, therapeutic cooling has been increasingly embraced by both tertiary medical centers and community hospitals. [40],[41],[42] However, a recent study comparing cooling of 33°C to 36°C in this patient population showed no difference in outcomes, [26] thus raising the issue of whether treatment of such patients should really focus on preventing hyperthermia. [43]

Therapeutic hypothermia has also been shown to be effective in preventing perinatal brain injury from HIE. There have been four clinical trials of newborns with HIE. [4],[6] These studies have shown benefit in infants with moderate and severe HIE; however, long-term, lifelong benefits are especially key in pediatric populations, and there are no reports of outcomes beyond 21 months of age. This condition has also been studied in the laboratory although not as extensively as in adult models. However, hypothermic protection in neonatal animal models has shown similar associations such as reduced excitatory amino acid accumulation, preservation of metabolic substrates, and inhibition of caspase activation. [44],[45]

Focal cerebral ischemia

In abundant studies of experimental stroke (focal cerebral ischemia), mild or moderate hypothermia has been shown to decrease infarct size and lead to functional improvement when cooling was initiated within a few hours of ischemia onset. Hypothermia reduced infarct volume and improved neurological function in the temperature range of 24°C to 33°C. [27],[28] The timing of hypothermia is important, as studies found that hypothermia initiated within 2~3 h of ischemia onset led to significant neuroprotection. Delays of 2~3 h were neuroprotective when cooling was maintained for either a few hours [46] or 48 h. [47] Recently, many experimental studies also demonstrated that reperfusion after ischemic stroke increases the likelihood of a good outcome. [28] In models of transient middle cerebral artery occlusion (tMCAO), hypothermia consistently showed neuroprotection, whereas in models of permanent MCAO (pMCAO), the results were conflicting. Thus, therapeutic hypothermia in conjunction with reperfusion strategies in clinical trial design becomes important. [12]

As therapeutic hypothermia was shown to be neuroprotective in cardiac arrest patients, there is hope that hypothermia may similarly reduce morbidity and mortality following ischemic stroke. Indeed, a few preclinical studies show that mild therapeutic hypothermia initiated during acute ischemic stork or after a delay reduces infarct size and mitigates functional impairment in a recent meta-analysis. [15],[37] In addition to these studies, another study showed feasibility and tolerability as well as regimens to prevent or reduce shivering in the awake stroke patient using intravascular cooling devices to cool acute ischemic stroke patients. [7],[8] In a recent randomized multicenter study, an endovascular cooling device was used in combination with the administration of a tissue plasminogen activator in acute stroke patients. Here, patients could be treated within 0-6 h of symptom onset followed by endovascular cooling to 33°C for 24 h. While the study was not designed to evaluate relative efficacy of hypothermia treatment, this regimen appeared well tolerated although there was an increased incidence of pneumonia among cooled patients. [11] However, these studies were all small and larger prospective studies have yet to be published.

Hypothermia in hemorrhagic stroke

Intracerebral hemorrhage (ICH)

ICH is a devastating stroke, and the resulting morbidity and mortality is much higher than ischemic stroke. While less studied compared to ischemic stroke, experimental studies have been directed at determining the pathophysiology of ICH and at identifying effective treatments that includes the study of hypothermia. A few reports have shown that hypothermia reduced brain edema, inflammation, and blood-brain barrier (BBB) disruption after intrastriatal thrombin injections [48] and following injection of autologous whole blood [49],[50],[51] but this was not as a consistent finding across laboratories. In fact, some laboratories have observed that histological and functional benefits are not consistently found [50] and one report described increased bleeding in the brain among cooled animals. [51] Although this appears to depend on the model, insult severity, and timing of treatment, another study of delayed mild hypothermia (48 h) after ICH failed to reduce the lesion size when started soon after collagenase-induced ICH, whereas treatment was delayed 12 h led to neuroprotection. [51] Although earlier cooling appears more favorable in brain ischemia, studies in brain hemorrhage models found increased bleeding with early cooling (delays to 12 h post collagenase injection) but some protection occurred when cooling was delayed 12-24 h. [52],[53] It is possible that hypothermia could affect critical procoagulant and thrombolytic systems and predispose to bleeding in the acute period or that hypothermia exacerbated complications of the initial increased blood pressure that occurs in this model. These findings bear further investigation to clarify the reasons for worsening in certain scenarios, and whether cooling might be detrimental if not applied in an optimal manner. At the clinical level, Kollmar et al. recently reported that 12 patients with large ICH were treated with hypothermia to 35°C for 10 days (initiated 3-12 h after symptoms onset) and these patients were compared to data from a local hemorrhage data bank. [54] In the hypothermia group, edema volume remained stable during 14 days, whereas edema significantly increased in the control group. However, larger controlled clinical trials of hypothermia in ICH are lacking.

Subarachnoid hemorrhage (SAH)

SAH is typically due to aneurysmal rupture, and hypothermia is often used intraoperatively during aneurysm repair. There are several animal model studies of SAH that showed that hypothermia exhibited neuroprotection. Mild hypothermia applied for 2 h led to improved posthemorrhagic neurological deficits and reduced intracranial pressure and postoperative weight gain at 1-7 days if cooling was delayed 3 h after SAH. [55] Another study demonstrated the neuroprotective effects of hypothermia on acute imaging changes after experimental SAH using diffusion-weighted imaging (DWI) and magnetic resonance spectroscopy (MRS). [56] The investigators established that hypothermia improved early development of cytotoxic edema, lactate accumulation, and general metabolic stress after SAH in the rat. The mechanisms underlying this protective effect have not been explored as extensively as in the brain ischemia models but one study showed that hypothermia interrupted the early expression of genes associated with cellular stress such as c-jun and heat shock protein (Hsp) 70. [57] At the clinical level, Muroi et al. assessed the anti-inflammatory effects of combining hypothermia and high-dose barbiturates. The inflammatory response in seven patients with this intervention showed decreased systemic and cerebrospinal fluid levels of interleukin (IL)-1β, IL-6, and leukocyte counts compared to a group of eight patients who received no intervention although cooling increased tumor necrosis factor alpha (TNF-α). [58]

However, in a large multicenter randomized study, mild intraoperative hypothermia during surgery for intracranial aneurysm did not lead to improved neurologic outcomes among patients with mild SAH. [59] As a follow-up to this study, more recent trials have focused on therapeutic hypothermia for patients with poor grade SAH. One clinical study conducted by Seule et al. evaluated the feasibility and safety of mild hypothermia in patients with poor grade SAH and increased intracranial pressure and/or cerebral vasospasm. [60] However, there were several complications among cooled patients, leading the investigators to conclude that prolonged systemic hypothermia might be considered a last-resort option for a carefully selected group of younger SAH patients with persistent intracranial hypertension and/or cerebral vasospasm. Thus, the clinical effectiveness of therapeutic cooling for SAH remains unclear.

Mechanisms of hypothermic protection in ischemic stroke

Metabolism and cerebral blood flow (CBF)

The neuroprotective properties of hypothermia have a profound effect on decreasing metabolic rate and reduce blood flow during ischemic stroke. [61] Hypothermia decreases brain oxygen consumption and glucose metabolism and on an average decreases brain oxygen consumption by approximately 5%/°C fall in body temperature in the range of 22~37°C, [62] and in anesthetized animals, oxygen consumption decreases linearly when brain temperature is lowered from 38°C to 18°C. [63] Hypothermia can interrupt downstream consequences of increased lactate production due to dependence on anaerobic metabolism and the development of acidosis by preserving the brain's metabolic stores. [1] Hypothermia conserves high-energy phosphate compounds such as adenosine triphosphate (ATP) and maintains tissue pH. This may be relative to the effect of hypothermia on brain metabolism that conserves tissue ATP levels. ATP is needed to maintain ion gradients and when these concentration gradients are disturbed, such as in the case of ischemic stroke, calcium influx occurs and leads to increased extracellular glutamate levels. [64] Several investigators also demonstrated that hypothermia significantly decreases the release of excitotoxic amino acids and subsequent calcium influx due to cerebral ischemia. [13],[65] Further, the glutamate receptor 2 (GluR2) subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor is thought to limit calcium influx. GluR2 suppression by ischemia is thought to increase calcium entry that in turn potentiates neuronal injury. Some investigators have hypothesized that one neuroprotective mechanism of hypothermia is that it could salvage neurons from delayed calcium influx by upregulating GluR 2 receptors. [66] In a recent work, our group has established that hypothermia decreases ischemia-induced upregulation of calcium influx through a newly characterized calcium-sensing receptor (CaSR). CaSR is thought to sense changes in extracellular calcium levels but also appears to reciprocally downregulate gamma-aminobutyric acid (GABA) receptors, thereby decreasing inhibitory tone. Cerebral ischemia increases expression of CaSR while inhibiting GABA-B-R1 expression, while mild hypothermia prevents this. [53] Thus, CaSR might be studied as a potential target for treatment of stroke and related conditions. Excitotoxic neurotransmitters are released early after ischemia onset, and it is well known that glutamate antagonists have a rather narrow temporal therapeutic window of 1-2 h even in models of temporary middle cerebral artery occlusion (MCAO). Since earlier cooling is superior to delayed cooling, this may explain some of the protective effect of hypothermia. However, when cooling is applied after glutamate is already released, neuroprotection is still observed, [67] and points to additional mechanisms of protection.

Hypothermia generally leads to reduced CBF. Under injury conditions such as stroke, CBF decreases linearly between 18 °C and 37°C, and CBF and brain metabolism are preserved at lower temperatures. Ischemic stroke leads to initial CBF decreases as a result of vessel occlusion but upon reperfusion, hyperemia occurs followed by a gradual decline in CBF that occurs over a period of hours. Mild hypothermia reduces this hyperemia and prevents the gradual CBF reduction during reperfusion. [68] However, the effects of hypothermia on this delayed CBF reduction are conflicting. Some reports showed that hypothermia increases CBF during ischemia, whereas others showed reduced or no effect on CBF. [1] Reasons for these discrepancies are unclear but could be explained by the dynamic nature of CBF, especially in the poststroke period and the different CBF assays used by different laboratories. Regardless of this, these observations indicate that these mechanisms do not fully explain the protective effect of hypothermia in experimental models.

Early molecular events

Hypothermia also influences the ischemic stress response including the induction of immediate early genes. [69] Several studies examined the effect of hypothermia on the stress response, namely, the expression of HSPs that are upregulated in response to a variety of cellular stress. Indeed, some investigators have shown that 70 kD inducible Hsp70 was upregulated by ischemic stroke model and was further increased by hypothermia although hypothermia itself did not induce Hsp70 in nonischemic model [70],[71] and this might be consistent with its neuroprotective properties. [72] However, other investigators have shown that hypothermia downregulates Hsp70 expression under similar condition, [73] while others have shown no influence of cooling on its expression. [74] Thus, it is unclear whether hypothermic neuroprotection is mediated through the stress response. Further, the significance of hypothermia's effect on the immediate early gene is unclear, as no subsequent studies have been carried out to establish causality.

MicroRNAs (miRNAs), a subset of noncoding RNAs, have been a topic of recent investigation in brain injury models. They have been shown to increase in expression as early as 2 h after ischemia onset. [75] These miRNAs are thought to play a role in silencing mRNA and they have been found to regulate a variety of signaling pathways. The exact roles of specific miRNAs remain under investigation but a report in a model of brain trauma showed that miRNA expression is affected by cooling. [76] In particular, a few miRNAs including miR-874 and miR-451 were decreased by cooling. These same investigators then overexpressed miR-34a, miR-451, and miR-874 in cultured neurons and found that these miRNAs led to increased cellular vulnerability to stretch injury, suggesting that one mechanism of hypothermic protection might be through the downregulation of these miRNAs. [77]

Apoptosis

Several studies have examined the influence of hypothermia on cell death via apoptosis. There are two main apoptotic pathways: the intrinsic pathway that occurs within the cell at the level of the mitochondria and the extrinsic pathway that is triggered via a cell surface receptor. [78] Hypothermia interrupts apoptosis through both pathways but whether cooling has any effect on neuron survival depends on whether apoptosis is occurring in a given model or para­digm. In models of global cerebral ischemia, hypothermia can interfere with the expression of Bcl-2 family members such as Bax and Bcl-2. Hypothermia has also been shown to reduce cytochrome c release and decrease caspase activation. Downstream of Bcl-2 family proteins, protein kinase C delta (PKC-delta) (a protein kinase C isoform) has been shown to contribute negatively to ischemic injury. [79] Caspase-3 leads to transport of PKC-delta from the cytosol to the mitochondria and nucleus, where it interacts with other molecules to induce apoptosis. [80] In contrast, a different isoform, epsilon-PKC is antiapoptotic, and is degraded by caspases. Hypothermia did not appear to alter overall levels of PKC-delta, [81] but it blocked its translocation to the mitochondria and the nucleus and stimulated the activity of epsilon-PKC after ischemia. [82]

Extrinsic apoptotic pathways also appear to be activated by brain ischemia. The most widely studied apoptosis-inducing receptor and ligand are Fas and Fas ligand (FasL), respectively. How FasL binds Fas is somewhat unclear, as many reports indicate that FasL must be present on the cell's surface in order to engage Fas but other reports indicate that FasL must first be cleaved from the surface by activated matrix metalloproteinases (MMPs) and solubilized. Hypothermia seems to pre­vent this cleavage, as levels of soluble FASL are decreased in cooled rodent brains, as are the levels of several MMPs. [83],[84] The decreased level of soluble FASL was also associ­ated with decreased caspase-8 activation. [85]

In models of more severe stroke (MCAO of 2 h or longer), hypothermia does not appear to affect Bcl-2 family members or caspase activation but prevents cytochrome c release. [86] These observations might be explained by a third apoptotic pathway that involves direct cell death by mitochondrial apoptosis-inducing factor (AIF) release. Hypothermia was shown to reduce apoptotic cell death in a more severe model of MCAO while suppressing AIF translocation. [87]

Other studies have also investigated other molecules implicated in apoptotic pathways that have been shown to be affected by hypothermia as well. Phosphatase and tensin homolog (PTEN) is a tumor suppressor molecule with proapoptotic functions. PTEN deletion has previously been shown to prevent ischemic brain injury. [88] However, PTEN phosphorylation leads to its deactivation and is normally decreased by brain ischemia. [89] Under conditions of neuroprotective hypothermia, phosphorylated PTEN levels remained unchanged but were decreased under nonneuroprotective hypothermia. [81] Thus, the deactivated form of this proapoptotic protein seems to correlate to neuroprotection.

Survival pathways

Several neurotrophic factors in the brain have been studied with regard to their therapeutic potential in acute ischemic stroke. These factors are involved in multiple neuronal cell processes such as synaptic function and plasticity and to sustain neuronal cell differentiation. In brain ischemia models, exogenous administration of one or more of these factors seemed to improve functional neurological outcome without necessarily affecting the lesion size. Hypothermia increased several of these factors including brain-derived neu­rotrophic factor (BDNF) after ischemic brain insults, [90],[91] glial-derived neurotrophic factor (GDNF) [92] and neurotrophin. [93] Hypothermia also activated extracellular signal-regulated kinase-1/2 (ERK1/2) phosphorylation, a downstream element of BDNF signaling. [91] In other cases, hypothermia has been reported to induce protective effects of ERK1/2. [94]

Studies of hypothermia on survival signals such as Bcl-2 and Akt have been reported. Hypothermia appears to upregulate Bcl-2, and promotes activation of Akt. [95],[96] After phosphorylation by phosphoinositol 3-kinase (PI3K), activated Akt phosphorylates (and thus inactivates) proapoptotic proteins such as glycogen synthase-3beta (GSK-3beta) and Bcl-xL/Bcl-2-associated death promoter (BAD). In a animal model of cerebral ischemia, hypothermia reduced infarct size by maintaining Akt activity. This effect disappeared when an Akt inhibitor was added to hypothermia. [96]

Interestingly, although hypothermia downregulates a majority of cell death pathways, it also upreglates several cell survival and growth pathways. Hypothermia has been shown to upregulate a family of cold shock protein such as cold-inducible RNA-binding protein (CIRP) and RNA-binding motif protein 3 (RBM3), [45],[97] that might be relevant to neuroprotection after ischemic stroke. CIRP has been speculated to protect and restore native RNA conformation during stress, and protects against apoptosis by upregulating ERK. RBM3 may the protect the brain from injury, as protection from cooling is no longer possible with gene knockdown in cultured neurons. [62] Further, in a model of Alzheimer's, RBM3 appeared to be important in synapse regeneration, and synapse loss could be prevented by cooling or RBM3 overexpression. [98] Using a model of apoptosis in the brain slices and neuron cultures, Chip et al. [99] showed that the cold shock protein RBM3 was induced by hypothermia, and its knockdown prevented any protective effect of cooling. In an in vitro model of neuronal apoptosis, CIRP overexpression led to the inhibition of apoptosis in a manner similar to that of hypothermia. [100] Thus, cold shock responses and corresponding proteins may prove to be important mechanisms underlying hypothermic protection, especially when such protection cannot be fully explained by salutary changes in brain metabolism, CBF, and excitotoxin release.

Inflammation

Inflammation is well-known to accompany a variety of acute neurological conditions such as stroke and other neuronal damage. Dead cells are known to trigger the activation of several immune responses via microglia and infiltrating immune cells from the circulation. [101] Further, there is evidence that brain cells not normally viewed as immunologic, including astrocytes and even neurons, can elaborate immune molecules. Several animal studies have now shown that inhibiting various aspects of this immune response by hypothermia may improve outcome following brain ischemia and injury. [102] Following ischemic stroke, inflammation can be detected within a few hours after the onset of the insult. Due to the acute nature of these brain insults, the ensuing immune response is most likely innate, rather than adaptive. The innate immune response is a triggered by signals that, unlike the adaptive immune response, do not require antigen recognition. Mild and moderate hypothermia influence several inflammatory response pathways. [89] Microglia play an important role in this innate response. [103] They display a ramified appearance while in the resting state but when activated, undergo a series of morphologic changes often leading to an amoeboid morphology. Microglial activation is the initial step in the CNS inflammatory response. Depending on the stimulus, this step may be followed by infiltration of circulating monocytes, neutrophils, and T cells, and by reactive astrocytosis. [104] Microglial activation is a complex series of events and the changes associated with microglial activation vary depending on the type, severity, and duration of the stimulus. [105] Hypothermia appears to decrease tissue density and the activation of microglia.

Endogenous immune stimulators, collectively referred to as damage-associated molecular patterns (DAMPs) are increased after ischemia. DAMPs include hyaluronan, surfactant protein, and uric acid. These substances can then bind to and stimulate receptors on microglia and other immune cells leading to the upregulation of many immune mediators through activation of several proinflammatory transcription factors including nuclear factor kappa B (NF-kB), [106] hypoxia-inducible factor 1 (HIF-1), interferon regulator factor 1, and signal transducer and activator of transcription 3 (STAT3). [107] Several studies showed that hypothermia also suppresses NF-kB, a transcription factor that activates many inflammation-related genes. NF-kB is normally present in the cytosol bound to its inhibitor protein IkB. When activated, IkB kinase (IKK) degrades IkB and releases NF-kB to enter the nucleus where it binds its consensus sequences. In focal cerebral ischemia, hypothermia inhibited IKK activity and prevented NF-kB translocation to the nucleus. [108] However, in a model of global ischemia, hypothermia appeared to associate with NF-kB, and IkB and IKK were unaffected. [109] Hypothermia also attenuated phosphorylation of STAT-3 that suppressed intercellular adhesion molecule-1 (ICAM-1) induction in the vascular system of stroke models. [110]

Cytokines were originally described as mediators involved in regulating the innate and adaptive immune systems. Cytokines are quickly and extensively upregulated in the brain in a variety of disease states. [101],[111] The most studied cytokines related to inflammation in acute brain injury are tumor necrosis factor-α (TNF-α), the ILs, especially IL-1, IL-4, IL-6, IL-10, and IL-18 and transforming growth factor (TGF)-β. Among these cytokines, mild hypothermia can reduce the expression levels of pro-inflammatory cytokine (IL-1β) and TNF- α in ischemic area. [96],[101] However, hypothermia also suppressed anti-inflammatory cytokines such as IL-10 and TGF-beta. [112]

Oxidative and nitrosative stresses may play a central role in this inflammatory response. ROS are also released by inflammatory cells. Studies have now shown that hypothermia attenuates ROS formation. [113] Similarly, nitrosative stresses include the increase of nitric oxide (NO) via the different nitric oxide synthase (NOS) isoforms, particularly the inducible NOS (iNOS) that is primarily found in immune cells. [114] In experimental stroke, hypothermia significantly attenuates increases in NOS isoforms [115] and subsequent NO generation. [116]

While hypothermia's effect on inflammation is largely suppressive, some immune signaling pathways may be important to stroke outcome. Recent work from our laboratory showed that while hypothermia largely suppressed microglial activation, selective upregulation of the novel innate immune receptor, triggering receptor expressed on myeloid cells-2 (TREM-2) was actually higher among microglia in brains protected by therapeutic cooling. [117] Deficiency of TREM-2 led to impaired phagocytosis and exacerbation of outcome in experimental stroke. [118] Thus, therapeutic cooling appears to differentially express immune molecules in such a way as to promote cell survival.

Blood-brain barrier and edema

Secondary injury due to BBB disruption leads to edema and hemorrhage. Studies have shown that mild and moderate hypothermia protects the BBB [119] and reduces edema formation [120] and attenuates loss of vascular basement proteins. [121] MMPs are proteases that break down the extracellular matrix, and disrupt the BBB leading to further infiltration of circulating immune cells, serum proteins and hemorrhage. [122] Inactivated MMPs are normally found in the cytosol, but in pathologic states, are transported extracellularly where they are cleaved to an active form and degrade substrates of the extracellular matrix. [101] MMP-2, -3, and -9 have been described in cerebral ischemia but MMP-9 appears to be expressed in traditional immune cells. Neutrophil MMP-9 expression after stroke correlates to worse outcome. [123] Studies using bone marrow chimeras suggest that MMP-9 derived from circulating leukocytes contributes significantly to stroke pathology. [124] Hypothermia reduces proteolytic activities of MMPs and consequent degradation of vascular basement membrane proteins [121] and the extracellular matrix. [52],[120],[125] Hypothermia also prevented the degradation of extracellular matrix proteins agrin and laminin, both targets of activated MMPs. [121] In addition to suppressing MMPs, hypothermia has been shown to increase expression of endogenous MMP inhibitors such as tissue inhibitor of metalloproteinase-2 (TIMP-2). [84]

Brain edema also results from upregulation of water channel proteins known as the aquaporins. Aquaporins facilitate water flux through the plasma membrane. In rodent brain, several studies have demonstrated the presence of different aquaporins. Among these aquaprins, aquaprin-4 (AQP4) is the predominant type of aquaporin in the microvasculature of the brain, present on astrocytic end-feet in contact with brain vessels. AQP4 expression is increased in reactive astrocytes in cerebral ischemic lesions, [95] and its deficiency has been shown to reduce brain edema following MCAO. [126] Mild hypothermia reduced brain edema formation by suppressing AQP4 expression in models of ICH [49] and cardiac arrest. [127]

Studies have also shown that hypothermia reduces brain hemorrhage which result from ischemic stroke (hemorrhagic transformation). [128] However, in models of protease-induced brain hemorrhage, hypothermia has resulted in conflicting results. Although several laboratories have shown neurological improvement in primary brain hemorrhage models, others reported no effect or even worsening outcomes. [120],[129] This may be because hypothermia causes a potential deficit in fibrinogen availability and a delay in thrombin generation, thereby inhibiting coagulation pathways. The net result of cooling may thus, lead to an increase in bleeding and exacerbation of hemorrhage. [130] Thus, hypothermia may be a less effective treatment in brain hemorrhage than in brain ischemia.

Recovery and repair

Studies investigating the long-term impact of hypothermia have observed trends after acute phases of injury and treatment, from weeks to months after cooling has ceased. Recent work concerning the question of hypothermia's lasting effects has specifically examined ongoing recovery and repair mechanisms after insults such as focal cerebral ischemia and traumatic brain injury. [131] Though the research has yet to reach a detailed consensus on the matter, studies have identified correlations between therapeutic hypothermia and the injured brain's regenerative capacity in stem-cell retention, neuronal synaptic connectivity repair, and neurogenesis as well as gliogenesis and angiogenesis.

Neurons in the injured brain are known to change morphology and lose synaptic connectivity as they undergo cell death. [132] More recent research has shown that, concurrent to neuronal loss, endogenous recovery mechanisms are also activated after injury, leading to some neurogenesis and synaptogenesis. Though neurogenesis appears rarely in the injured brain, [133] rodent studies have shown that acute brain insults initiate the proliferation of neural stem cells in the subventricular and subgranular zones. [132] However, spontaneous recovery by neurogenesis is limited in brain injury, and there is an obvious need to develop strategies to improve regenerative processes including the proliferation of neuronal precursor cells, migration of precursor cells to the injury area, differentiation into mature neurons, and reconnection between neurons. [134]

To date, the relationship between hypothermia and neurogenesis has only been studied by a few groups and is far from clear. Studies examining mild hypothermia in cultured neural stem cells showed decreased apoptosis, an increase in nestin-positive cells, and inhibition of stem cell differentiation into astrocytes, [135] suggesting an overall inductive role for hypothermia in neurogenesis. However, the effects of hypothermia on neurogenesis have also been shown to vary according to conditions such as age, injured versus noninjured state, and the severity or duration of hypothermia. One study in the developing brain showed that cooling to 30°C for 21 h decreased the number of proliferating cells in the subgranular zone of the hippocampus but not in the periventricular zone. [136] After hypoxic-ischemic injury, cooling the developing brain to 33°C showed an increase in neural progenitor cell differentiation in the striatum as well as protection of proliferating neural stem cells that had been produced in response to ischemic stimuli. [137] This pattern of reduced neural stem cell apoptosis has been associated with hypothermia-induced increases in the expression of Bcl-2. Meanwhile, forebrain ischemia studies in adult rodents showed that mild hypothermia increased neurogenesis in the dentate gyrus relative to similarly injured normothermic animals. [138] Another study involving adult rats and forebrain ischemia showed that cooling after injury had no effect on neurogenesis. [139] This second study employed a similar model of forebrain ischemia but a shorter window of hypothermia (33°C for 45 min) that was applied either in the acute and subacute phases of stroke, whether during injury or the immediate reperfusion phase. [140] The findings in the adult brain suggest that hypothermia can only influence neurogenesis within time windows that remain poorly defined.

Reports of hypothermia's effects on endogenous cell genesis in injured brains relative to uninjured brains have been inconsistent. There are conflicting reports as to whether hypothermia suppresses stem cell proliferation [136],[141] or induces it; [135],[138] some even suggest that hypothermia may preferentially promote cell differentiation toward neurogenesis over gliogenesis. Studies also indicate that the effect of hypothermia on gliogenesis is dependent on cooling temperature. Hypothermia to temperatures lower than 30°C has been shown to induce apoptosis/necrosis and cell cycle arrest as a result of reduced energy supply, thereby suppressing cell proliferation. [136],[142] On the contrary, mild hypothermia has shown to be protective against progenitor cell death. [135],[141]

Brain injury studies have observed astrogliogenesis and angiogenesis contributing to brain recovery following insult. [132],[143] As of yet, the role of newborn astrocytes or endothelial cells in the brain has not been studied extensively. Astrocytes comprise the largest population of cells in the ischemic core following the acute period of stroke. [144] Glial scarring is thought to obstruct new neurite outgrowth. [145],[146] However, the inhibition of astrocytic activation can exacerbate injury responses. [146] As for angiogenesis, mild hypothermia has been observed to increase angiogenic signals in focal ischemia, [147] spinal cord injury, [148] and traumatic brain injury models [149] but the research has yet to qualify the significance of these trends. In fact, some of the research suggests that angiogenesis may be detrimental to brain repair. One clinical study measured the levels of angiogenic factors in samples from acute stroke patients including platelet-derived growth factors (PDGFs), vascular endothelial growth factors (VEGFs) and their receptors, stromal cell-derived factor 1 (SDF-1), and hepatocyte growth factor (HGF). The study concluded that acute antiangiogenic status predicted worse long-term functional outcomes. However, the data also showed that an early predominance of proangiogenic factors is associated with milder short-term neurological deficits. [150]

Though less well-understood, oligodendrocytes are known to respond to brain injury in a manner similar to neurons. Hypothermia has also been shown to attenuate trauma-induced oligodendrocyte death, demyelination, and circuit disruption. [151] Hypothermia improved survival in primary cultures of mouse oligodendrocyte precursor cells, [152] demonstrating that cooling can help the prenatal brain retain a greater number of actively replicating, less differentiated oligodendrocyte precursor cells. Meanwhile, an in vivo study of fetal sheep in utero exposed to hypoxic conditions showed that hypothermia (30°C) reduced overall immature oligodedrocyte loss but did not prevent injury-induced decreases in proliferating oligodendrocytes within the periventricular white matter.

Few studies have examined the role of hypothermia on neuronal circuit repair. After stem cell protection and proliferation, repair of synaptic connectivity is crucial to functional recovery after brain injury. Deep hypothermia (17C) showed neurite and axonal outgrowth in brain slices, [94],[153] suggesting that cooling may have a restorative effect on cell morphology. Mild hypothermia has also been shown to significantly alter hipppocampal gene expression in rat brains after traumatic brain injury. One study identified 133 transcripts altered by brain injury, the expression profiles of which were statistically different between the hypothermic and normothermic groups. Of the 57 transcripts that were upregulated by hypothermia, prominent increases were observed among genes related to synapse organization and biogenesis. While the full effect of cooling in brain repair is still unclear, the current research suggests that therapeutic hypothermia may have a beneficial role under specific conditions, whether by protecting stem cells, promoting their proliferation and differentiation, increasing growth factor signaling, or encouraging the recovery of neural circuitry.


  Conclusions Top


Hypothermia has long been known to be a potent neuroprotective intervention that preserves tissues and limits injury after ischemic stroke. Experimental evidence and clinical experience show that induced hypothermia affects nearly every metabolic, molecular, and cellular event in cell death to promote tissue preservation. More recent insights suggest that hypothermia can also favorably modulate endogenous regenerative and restorative properties. The many ways therapeutic hypothermia effects protection has shown that the goal of neuroprotection requires multitarget approaches after acute ischemic stroke.

It may also be possible to extend the therapeutic window for other neuroprotective treatments by hypothermia, and combination therapies with neuroprotective drugs such as anti-inflammatory and thrombolytic agents. This effect of hypothermia may lead to the reexamination of the many failed neuroprotectant drugs at the clinical level since many drugs may not have been studied under optimal conditions. In spite of the robust protective effect demonstrated in the laboratory, there are still clinical obstacles to overcome including effective cooling in humans, prevention of harmful side-effects, and identifying patient populations most likely to benefit. There is also a need to develop more sophisticated translational research tools in the laboratory. Animal models and the method of cooling used in the laboratory are quite different from those employed clinically. Thus, an effort to simulate the clinical condition more precisely might provide solutions for better and wider application of therapeutic hypothermia in human patients. Second, there are few investigations into overcoming the complications of systemic hypothermia such as shivering, infection, and coagulopathies. Though these complications are largely ignored in the laboratory, they are significant at the clinical level and will need to be addressed.

As such, it seems wise to approach stroke therapy with combination therapies including neuroprotective, anti-inflammatory, and antiapoptotic treatments plus recanalization. In essence, the usage of combination therapies do all that cooling does without the risks.

Acknowledgements

This work was supported by grants from the National Institutes of Health (NS40516), and the Veteran's Merit Award to MY and an American Heart Association Western States Affiliate Postdoctoral Fellowship (13POST14810019) to JYK. These grants were administered by the Northern California Institute for Research and Education and supported by resources of the Veterans Affairs Medical Center, San Francisco, California, USA.

Financial support and sponsorship

NIH, AHA, VA merit.

Conflicts of interest

There are no conflicts of interest.

 
  References Top

1.
Yenari MA, Han HS. Neuroprotective mechanisms of hypothermia in brain ischaemia. Nat Rev Neurosci 2012;13:267-78.   Back to cited text no. 1
    
2.
Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G, et al. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med 2002;346:557-63.   Back to cited text no. 2
    
3.
Hypothermia after Cardiac Arrest Study Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med 2002;346:549-56.   Back to cited text no. 3
    
4.
Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA, Donovan EF, et al.; National Institute of Child Health and Human Development Neonatal Research Network. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med 2005;353:1574-84.   Back to cited text no. 4
    
5.
Gluckman PD, Wyatt JS, Azzopardi D, Ballard R, Edwards AD, Ferriero DM, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: Multicentre randomised trial. Lancet 2005;365:663-70.   Back to cited text no. 5
    
6.
Azzopardi DV, Strohm B, Edwards AD, Dyet L, Halliday HL, Juszczak E, et al.; TOBY Study Group. Moderate hypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med 2009;361:1349-58.   Back to cited text no. 6
    
7.
Lyden PD, Allgren RL, Ng K, Akins P, Meyer B, Al-Sanani F, et al. Intravascular Cooling in the Treatment of Stroke (ICTuS): Early clinical experience. J Stroke Cerebrovasc Dis 2005;14:107-14.   Back to cited text no. 7
    
8.
De Georgia MA, Krieger DW, Abou-Chebl A, Devlin TG, Jauss M, Davis SM, et al. Cooling for acute ischemic brain damage (COOL AID): A feasibility trial of endovascular cooling. Neurology 2004;63:312-7.   Back to cited text no. 8
    
9.
Schwab S, Schwarz S, Spranger M, Keller E, Bertram M, Hacke W. Moderate hypothermia in the treatment of patients with severe middle cerebral artery infarction. Stroke 1998;29:2461-6.   Back to cited text no. 9
    
10.
Schwab S, Georgiadis D, Berrouschot J, Schellinger PD, Graffagnino C, Mayer SA. Feasibility and safety of moderate hypothermia after massive hemispheric infarction. Stroke 2001;32:2033-5.   Back to cited text no. 10
    
11.
Hemmen TM, Raman R, Guluma KZ, Meyer BC, Gomes JA, Cruz-Flores S, et al.; ICTuS-L Investigators. Intravenous thrombolysis plus hypothermia for acute treatment of ischemic stroke (ICTuS-L): Final results. Stroke 2010;41:2265-70.   Back to cited text no. 11
    
12.
Lyden PD, Krieger D, Yenari M, Dietrich WD. Therapeutic hypothermia for acute stroke. Int J Stroke 2006;1:9-19.   Back to cited text no. 12
    
13.
Busto R, Dietrich WD, Globus MY, Ginsberg MD. The importance of brain temperature in cerebral ischemic injury. Stroke 1989;20:1113-4.   Back to cited text no. 13
    
14.
Busto R, Dietrich WD, Globus MY, Valdés I, Scheinberg P, Ginsberg MD. Small differences in intraischemic brain temperature critically determine the extent of ischemic neuronal injury. J Cereb Blood Flow Metab 1987;7:729-38.   Back to cited text no. 14
    
15.
van der Worp HB, Macleod MR, Kollmar R; European Stroke Research Network for Hypothermia (EuroHYP). Therapeutic hypothermia for acute ischemic stroke: Ready to start large randomized trials? J Cereb Blood Flow Metab 2010;30:1079-93.   Back to cited text no. 15
    
16.
Huh PW, Belayev L, Zhao W, Koch S, Busto R, Ginsberg MD. Comparative neuroprotective efficacy of prolonged moderate intraischemic and postischemic hypothermia in focal cerebral ischemia. J Neurosurg 2000;92:91-9.   Back to cited text no. 16
    
17.
Maier CM, Ahern Kv, Cheng ML, Lee JE, Yenari MA, Steinberg GK. Optimal depth and duration of mild hypothermia in a focal model of transient cerebral ischemia: Effects on neurologic outcome, infarct size, apoptosis, and inflammation. Stroke 1998;29:2171-80.   Back to cited text no. 17
    
18.
Kollmar R, Blank T, Han JL, Georgiadis D, Schwab S. Different degrees of hypothermia after experimental stroke: Short- and long-term outcome. Stroke 2007;38:1585-9.   Back to cited text no. 18
    
19.
Krieger DW, De Georgia MA, Abou-Chebl A, Andrefsky JC, Sila CA, Katzan IL, et al. Cooling for acute ischemic brain damage (cool aid): An open pilot study of induced hypothermia in acute ischemic stroke. Stroke 2001;32:1847-54.   Back to cited text no. 19
    
20.
Jacobs S, Hunt R, Tarnow-Mordi W, Inder T, Davis P. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2007:CD003311.   Back to cited text no. 20
    
21.
Hutchison JS, Doherty DR, Orlowski JP, Kissoon N. Hypothermia therapy for cardiac arrest in pediatric patients. Pediatr Clin North Am 2008;55:529-44, ix.   Back to cited text no. 21
    
22.
Peterson K, Carson S, Carney N. Hypothermia treatment for traumatic brain injury: A systematic review and meta-analysis. J Neurotrauma 2008;25:62-71.   Back to cited text no. 22
    
23.
Polderman KH. Mechanisms of action, physiological effects, and complications of hypothermia. Crit Care Med 2009;37(Suppl):S186-202.   Back to cited text no. 23
    
24.
Tokutomi T, Miyagi T, Takeuchi Y, Karukaya T, Katsuki H, Shigemori M. Effect of 35 degrees C hypothermia on intracranial pressure and clinical outcome in patients with severe traumatic brain injury. J Trauma 2009;66:166-73.   Back to cited text no. 24
    
25.
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;31:2251-6.   Back to cited text no. 25
    
26.
Nielsen N, Wetterslev J, Cronberg T, Erlinge D, Gasche Y, Hassager C, et al.; TTM Trial Investigators. Targeted temperature management at 33 ° C versus 36 ° C after cardiac arrest. N Engl J Med 2013;369:2197-206.   Back to cited text no. 26
    
27.
Clark DL, Penner M, Orellana-Jordan IM, Colbourne F. Comparison of 12, 24 and 48 h of systemic hypothermia on outcome after permanent focal ischemia in rat. Exp Neurol 2008;212:386-92.   Back to cited text no. 27
    
28.
Krieger DW, Yenari MA. Therapeutic hypothermia for acute ischemic stroke: What do laboratory studies teach us? Stroke 2004;35:1482-9.   Back to cited text no. 28
    
29.
Colbourne F, Li H, Buchan AM. Indefatigable CA1 sector neuroprotection with mild hypothermia induced 6 hours after severe forebrain ischemia in rats. J Cereb Blood Flow Metab 1999;19:742-9.   Back to cited text no. 29
    
30.
Lawrence EJ, Dentcheva E, Curtis KM, Roberts VL, Siman R, Neumar RW. Neuroprotection with delayed initiation of prolonged hypothermia after in vitro transient global brain ischemia. Resuscitation 2005;64:383-8.   Back to cited text no. 30
    
31.
Zhang ZG, Chopp M, Chen H. Duration dependent post-ischemic hypothermia alleviates cortical damage after transient middle cerebral artery occlusion in the rat. J Neurol Sci 1993;117:240-4.   Back to cited text no. 31
    
32.
Carroll M, Beek O. Protection against hippocampal CA1 cell loss by post-ischemic hypothermia is dependent on delay of initiation and duration. Metab Brain Dis 1992;7:45-50.   Back to cited text no. 32
    
33.
Dietrich WD, Busto R, Alonso O, Globus MY, Ginsberg MD. Intraischemic but not postischemic brain hypothermia protects chronically following global forebrain ischemia in rats. J Cereb Blood Flow Metab 1993;13:541-9.   Back to cited text no. 33
    
34.
Shuaib A, Waqar T, Wishart T, Kanthan R. Post-ischemic therapy with CGS-19755 (alone or in combination with hypothermia) in gerbils. Neurosci Lett 1995;191:87-90.   Back to cited text no. 34
    
35.
Colbourne F, Corbett D. Delayed postischemic hypothermia: A six month survival study using behavioral and histological assessments of neuroprotection. J Neurosci 1995;15:7250-60.   Back to cited text no. 35
    
36.
Olsen TS, Weber UJ, Kammersgaard LP. Therapeutic hypothermia for acute stroke. Lancet Neurol 2003;2:410-6.   Back to cited text no. 36
    
37.
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;130:3063-74.   Back to cited text no. 37
    
38.
Meloni BP, Mastaglia FL, Knuckey NW. Therapeutic applications of hypothermia in cerebral ischaemia. Ther Adv Neurol Disord 2008;1:12-35.   Back to cited text no. 38
    
39.
Colbourne F, Corbett D. Delayed and prolonged post-ischemic hypothermia is neuroprotective in the gerbil. Brain Res 1994;654:265-72.   Back to cited text no. 39
    
40.
Arrich J, Holzer M, Havel C, Müllner M, Herkner H. Hypothermia for neuroprotection in adults after cardiopulmonary resuscitation. Cochrane Database Syst Rev 2012;9:CD004128.   Back to cited text no. 40
    
41.
Fink EL, Clark RS, Kochanek PM, Bell MJ, Watson RS. A tertiary care center's experience with therapeutic hypothermia after pediatric cardiac arrest. Pediatr Crit Care Med 2010;11:66-74.   Back to cited text no. 41
    
42.
Shah MP, Zimmerman L, Bullard J, Yenari MA. Therapeutic hypothermia after cardiac arrest: Experience at an academically affiliated community-based veterans affairs medical center. Stroke Res Treat 2011;2011:791639.   Back to cited text no. 42
    
43.
Kim JY, Kim N, Yenari MA, Chang W. Hypothermia and pharmacological regimens that prevent overexpression and overactivity of the extracellular calcium-sensing receptor protect neurons against traumatic brain injury. J Neurotrauma 2013;30:1170-6.   Back to cited text no. 43
    
44.
Laptook AR. Use of therapeutic hypothermia for term infants with hypoxic-ischemic encephalopathy. Pediatr Clin North Am 2009;56:601-16, Table of Contents.   Back to cited text no. 44
    
45.
Tang XN, Yenari MA. Hypothermia as a cytoprotective strategy in ischemic tissue injury. Ageing Res Rev 2010;9:61-8.   Back to cited text no. 45
    
46.
Maier CM, Sun GH, Kunis D, Yenari MA, Steinberg GK. Delayed induction and long-term effects of mild hypothermia in a focal model of transient cerebral ischemia: Neurological outcome and infarct size. J Neurosurg 2001;94:90-6.   Back to cited text no. 46
    
47.
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. J Cereb Blood Flow Metab 2000;20:1702-8.   Back to cited text no. 47
    
48.
Kawai N, Kawanishi M, Okauchi M, Nagao S. Effects of hypothermia on thrombin-induced brain edema formation. Brain Res 2001;895:50-8.   Back to cited text no. 48
    
49.
Dai DW, Wang DS, Li KS, Mao Y, Zhang LM, Duan SR, et al. Effect of local mild hypothermia on expression of aquaporin-4 following intracerebral hemorrhage in rats. Zhonghua Yi Xue Za Zhi 2006;86:906-10.   Back to cited text no. 49
    
50.
Fingas M, Clark DL, Colbourne F. The effects of selective brain hypothermia on intracerebral hemorrhage in rats. Exp Neurol 2007;208:277-84.   Back to cited text no. 50
    
51.
MacLellan CL, Davies LM, Fingas MS, Colbourne F. The influence of hypothermia on outcome after intracerebral hemorrhage in rats. Stroke 2006;37:1266-70.   Back to cited text no. 51
    
52.
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 Neuropathol 2004;108:406-12.   Back to cited text no. 52
    
53.
Kim JY, Kim N, Yenari MA, Chang W. Mild hypothermia suppresses calcium-sensing receptor (CaSR) induction following forebrain ischemia while increasing GABA-B receptor 1 (GABA-B-R1) expression. Transl Stroke Res 2011;2:195-201.   Back to cited text no. 53
    
54.
Kollmar R, Staykov D, Dorfler A, Schellinger PD, Schwab S, Bardutzky J. Hypothermia reduces perihemorrhagic edema after intracerebral hemorrhage. Stroke 2010;41:1684-9.   Back to cited text no. 54
    
55.
Torok E, Klopotowski M, Trabold R, Thal SC, Plesnila N, Schöller K. Mild hypothermia (33 degrees C) reduces intracranial hypertension and improves functional outcome after subarachnoid hemorrhage in rats. Neurosurgery 2009;65:352-9.  Back to cited text no. 55
    
56.
Schubert GA, Poli S, Mendelowitsch A, Schilling L, Thomé C. Hypothermia reduces early hypoperfusion and metabolic alterations during the acute phase of massive subarachnoid hemorrhage: A laser-Doppler-flowmetry and microdialysis study in rats. J Neurotrauma 2008;25:539-48.   Back to cited text no. 56
    
57.
Kawamura Y, Yamada K, Masago A, Katano H, Matsumoto T, Mase M. Hypothermia modulates induction of hsp70 and c-jun mRNA in the rat brain after subarachnoid hemorrhage. J Neurotrauma 2000;17:243-50.   Back to cited text no. 57
    
58.
Muroi C, Frei K, El Beltagy M, Cesnulis E, Yonekawa Y, Keller E. Combined therapeutic hypothermia and barbiturate coma reduces interleukin-6 in the cerebrospinal fluid after aneurysmal subarachnoid hemorrhage. J Neurosurg Anesthesiol 2008;20:193-8.   Back to cited text no. 58
    
59.
Todd MM, Hindman BJ, Clarke WR, Torner JC; Intraoperative Hypothermia for Aneurysm Surgery Trial (IHAST) Investigators. Mild intraoperative hypothermia during surgery for intracranial aneurysm. N Engl J Med 2005;352:135-45.   Back to cited text no. 59
    
60.
Seule MA, Muroi C, Mink S, Yonekawa Y, Keller E. Therapeutic hypothermia in patients with aneurysmal subarachnoid hemorrhage, refractory intracranial hypertension, or cerebral vasospasm. Neurosurgery 2009;64:86-93.   Back to cited text no. 60
    
61.
Shackelford RT, Hegedus SA. Factors affecting cerebral blood flow--experimental review: Sympathectomy, hypothermia, CO2 inhalation and pavarine. Ann Surg 1966;163:771-7.   Back to cited text no. 61
    
62.
Yenari MA, Wijman CA, Steinberg GK. Effects of hypothermia on cerebral metabolism, blood flow and autoregulation. In: Mayer S, Sessler DI, editors. Therapeutic Hypothermia. NY, NY: Marcel Dekker Inc; 2004. p. 141-78.  Back to cited text no. 62
    
63.
Ehrlich MP, McCullough JN, Zhang N, Weisz DJ, Juvonen T, Bodian CA, et al. Effect of hypothermia on cerebral blood flow and metabolism in the pig. Ann Thorac Surg 2002;73:191-7.   Back to cited text no. 63
    
64.
Lee JM, Zipfel GJ, Choi DW. The changing landscape of ischaemic brain injury mechanisms. Nature 1999;399(Suppl):A7-14.   Back to cited text no. 64
    
65.
Matsumoto M, Scheller MS, Zornow MH, Strnat MA. Effect of S-emopamil, nimodipine, and mild hypothermia on hippocampal glutamate concentrations after repeated cerebral ischemia in rabbits. Stroke 1993;24:1228-34.   Back to cited text no. 65
    
66.
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. Proc Natl Acad Sci U S A 2003;100:2906-10.   Back to cited text no. 66
    
67.
Ginsberg MD, Sternau LL, Globus MY, Dietrich WD, Busto R. Therapeutic modulation of brain temperature: Relevance to ischemic brain injury. Cerebrovasc Brain Metab Rev 1992;4:189-225.   Back to cited text no. 67
    
68.
Yenari M, Kitagawa K, Lyden P, Perez-Pinzon M. Metabolic downregulation: A key to successful neuroprotection? Stroke 2008;39:2910-7.   Back to cited text no. 68
    
69.
Kamme F, Campbell K, Wieloch T. Biphasic expression of the fos and jun families of transcription factors following transient forebrain ischaemia in the rat. Effect of hypothermia. Eur J Neurosci 1995;7:2007-16.   Back to cited text no. 69
    
70.
Terao Y, Miyamoto S, Hirai K, Kamiguchi H, Ohta H, Shimojo M, et al. Hypothermia enhances heat-shock protein 70 production in ischemic brains. Neuroreport 2009;20:745-9.   Back to cited text no. 70
    
71.
Cullen KE, Sarge KD. Characterization of hypothermia-induced cellular stress response in mouse tissues. J Biol Chem 1997;272:1742-6.   Back to cited text no. 71
    
72.
Yenari MA, Liu J, Zheng Z, Vexler ZS, Lee JE, Giffard RG. Antiapoptotic and anti-inflammatory mechanisms of heat-shock protein protection. Ann N Y Acad Sci 2005;1053:74-83.   Back to cited text no. 72
    
73.
Kumar K, Wu X, Evans AT, Marcoux F. The effect of hypothermia on induction of heat shock protein (HSP)-72 in ischemic brain. Metab Brain Dis 1995;10:283-91.   Back to cited text no. 73
    
74.
Xu L, Yenari MA, Steinberg GK, Giffard RG. Mild hypothermia reduces apoptosis of mouse neurons in vitro early in the cascade. J Cereb Blood Flow Metab 2002;22:21-8.   Back to cited text no. 74
    
75.
Vemuganti R. The MicroRNAs and stroke: No need to be coded to be counted. Transl Stroke Res 2010;1:158-60.   Back to cited text no. 75
    
76.
Truettner JS, Alonso OF, Bramlett HM, Dietrich WD. Therapeutic hypothermia alters microRNA responses to traumatic brain injury in rats. J Cereb Blood Flow Metab 2011;31:1897-907.   Back to cited text no. 76
    
77.
Truettner JS, Motti D, Dietrich WD. MicroRNA overexpression increases cortical neuronal vulnerability to injury. Brain Res 2013;1533:122-30.   Back to cited text no. 77
    
78.
Okouchi M, Ekshyyan O, Maracine M, Aw TY. Neuronal apoptosis in neurodegeneration. Antioxid Redox Signal 2007;9:1059-96.   Back to cited text no. 78
    
79.
Bright R, Raval AP, Dembner JM, Pérez-Pinzón MA, Steinberg GK, Yenari MA, et al. Protein kinase C delta mediates cerebral reperfusion injury in vivo. J Neurosci 2004;24:6880-8.  Back to cited text no. 79
    
80.
Raval AP, Dave KR, Prado R, Katz LM, Busto R, Sick TJ, et al. Protein kinase C delta cleavage initiates an aberrant signal transduction pathway after cardiac arrest and oxygen glucose deprivation. J Cereb Blood Flow Metab 2005;25:730-41.   Back to cited text no. 80
    
81.
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. J Cereb Blood Flow Metab 2009;29:1589-600.   Back to cited text no. 81
    
82.
Shimohata T, Zhao H, Steinberg GK. Epsilon PKC may contribute to the protective effect of hypothermia in a rat focal cerebral ischemia model. Stroke 2007;38:375-80.   Back to cited text no. 82
    
83.
Hamann GF, Burggraf D, Martens HK, Liebetrau M, Jäger G, Wunderlich N, et al. Mild to moderate hypothermia prevents microvascular basal lamina antigen loss in experimental focal cerebral ischemia. Stroke 2004;35:764-9.   Back to cited text no. 83
    
84.
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. J Neurosurg 2005;103:289-97.   Back to cited text no. 84
    
85.
Liu L, Kim JY, Koike MA, Yoon YJ, Tang XN, Ma H, et al. FasL shedding is reduced by hypothermia in experimental stroke. J Neurochem 2008;106:541-50.   Back to cited text no. 85
    
86.
Yenari MA, Iwayama S, Cheng D, Sun GH, Fujimura M, Morita-Fujimura Y, et al. Mild hypothermia attenuates cytochrome c release but does not alter Bcl-2 expression or caspase activation after experimental stroke. J Cereb Blood Flow Metab 2002;22:29-38.   Back to cited text no. 86
    
87.
Zhao H, Wang JQ, Shimohata T, Sun G, Yenari MA, Sapolsky RM, et al. Conditions of protection by hypothermia and effects on apoptotic pathways in a rat model of permanent middle cerebral artery occlusion. J Neurosurg 2007;107:636-41.   Back to cited text no. 87
    
88.
Shi GD, OuYang YP, Shi JG, Liu Y, Yuan W, Jia LS. PTEN deletion prevents ischemic brain injury by activating the mTOR signaling pathway. Biochem Biophys Res Commun 2011;404:941-5.   Back to cited text no. 88
    
89.
Zhao H, Steinberg GK, Sapolsky RM. General versus specific actions of mild-moderate hypothermia in attenuating cerebral ischemic damage. J Cereb Blood Flow Metab 2007; 27:1879-94.   Back to cited text no. 89
    
90.
Vosler PS, Logue ES, Repine MJ, Callaway CW. Delayed hypothermia preferentially increases expression of brain-derived neurotrophic factor exon III in rat hippocampus after asphyxial cardiac arrest. Brain Res Mol Brain Res 2005;135:21-9.   Back to cited text no. 90
    
91.
D'Cruz BJ, Fertig KC, Filiano AJ, Hicks SD, DeFranco DB, Callaway CW. Hypothermic reperfusion after cardiac arrest augments brain-derived neurotrophic factor activation. J Cereb Blood Flow Metab 2002;22:843-51.   Back to cited text no. 91
    
92.
Schmidt KM, Repine MJ, Hicks SD, DeFranco DB, Callaway CW. Regional changes in glial cell line-derived neurotrophic factor after cardiac arrest and hypothermia in rats. Neurosci Lett 2004;368:135-9.  Back to cited text no. 92
    
93.
Boris-Möller F, Kamme F, Wieloch T. The effect of hypothermia on the expression of neurotrophin mRNA in the hippocampus following transient cerebral ischemia in the rat. Brain Res Mol Brain Res 1998;63:163-73.   Back to cited text no. 93
    
94.
Schmitt KR, Diestel A, Lehnardt S, Schwartlander R, Lange PE, Berger F, et al. Hypothermia suppresses inflammation via ERK signaling pathway in stimulated microglial cells. J Neuroimmunol 2007;189:7-16.   Back to cited text no. 94
    
95.
Slikker W 3 rd , Desai VG, Duhart H, Feuers R, Imam SZ. Hypothermia enhances bcl-2 expression and protects against oxidative stress-induced cell death in Chinese hamster ovary cells. Free Radic Biol Med 2001;31:405-11.   Back to cited text no. 95
    
96.
Zhao H, Shimohata T, Wang JQ, Sun G, Schaal DW, Sapolsky RM, et al. Akt contributes to neuroprotection by hypothermia against cerebral ischemia in rats. J Neurosci 2005;25:9794-806.   Back to cited text no. 96
    
97.
Tong G, Endersfelder S, Rosenthal LM, Wollersheim S, Sauer IM, Bührer C, et al. Effects of moderate and deep hypothermia on RNA-binding proteins RBM3 and CIRP expressions in murine hippocampal brain slices. Brain Res 2013;1504:74-84.   Back to cited text no. 97
    
98.
Peretti D, Bastide A, Radford H, Verity N, Molloy C, Martin MG, et al. RBM3 mediates structural plasticity and protective effects of cooling in neurodegeneration. Nature 2015;518:236-9.   Back to cited text no. 98
    
99.
Chip S, Zelmer A, Ogunshola OO, Felderhoff-Mueser U, Nitsch C, Bührer C, et al. The RNA-binding protein RBM3 is involved in hypothermia induced neuroprotection. Neurobiol Dis 2011;43:388-96.   Back to cited text no. 99
    
100.
Zhang HT, Xue JH, Zhang ZW, Kong HB, Liu AJ, Li SC, et al. Cold-inducible RNA-binding protein inhibits neuron apoptosis through the suppression of mitochondrial apoptosis. Brain Res 2015. [Epub ahead of print].  Back to cited text no. 100
    
101.
Wang Q, Tang XN, Yenari MA. The inflammatory response in stroke. J Neuroimmunol 2007;184:53-68.   Back to cited text no. 101
    
102.
Ceulemans AG, Zgavc T, Kooijman R, Hachimi-Idrissi S, Sarre S, Michotte Y. The dual role of the neuroinflammatory response after ischemic stroke: Modulatory effects of hypothermia. J Neuroinflammation 2010;7:74.   Back to cited text no. 102
    
103.
Rivest S. Regulation of innate immune responses in the brain. Nat Rev Immunol 2009;9:429-39.   Back to cited text no. 103
    
104.
Zheng Z, Yenari MA. Post-ischemic inflammation: Molecular mechanisms and therapeutic implications. Neurol Res 2004;26:884-92.   Back to cited text no. 104
    
105.
Ransohoff RM. Immunology: Barrier to electrical storms. Nature 2009;457:155-6.   Back to cited text no. 105
    
106.
Ghosh S, May MJ, Kopp EB. NF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responses. Annu Rev Immunol 1998;16:225-60.   Back to cited text no. 106
    
107.
Yilmaz G, Granger DN. Cell adhesion molecules and ischemic stroke. Neurol Res 2008;30:783-93.   Back to cited text no. 107
    
108.
Han HS, Karabiyikoglu M, Kelly S, Sobel RA, Yenari MA. Mild hypothermia inhibits nuclear factor-kappaB translocation in experimental stroke. J Cereb Blood Flow Metab 2003;23:589-98.   Back to cited text no. 108
    
109.
Webster CM, Kelly S, Koike MA, Chock VY, Giffard RG, Yenari MA. Inflammation and NFkappaB activation is decreased by hypothermia following global cerebral ischemia. Neurobiol Dis 2009;33:301-12.   Back to cited text no. 109
    
110.
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 Res Treat 2011;2011:846716.   Back to cited text no. 110
    
111.
Trendelenburg G. Acute neurodegeneration and the inflammasome: Central processor for danger signals and the inflammatory response? J Cereb Blood Flow Metab 2008;28:867-81.   Back to cited text no. 111
    
112.
Matsui T, Kakeda T. IL-10 production is reduced by hypothermia but augmented by hyperthermia in rat microglia. J Neurotrauma 2008;25:709-15.   Back to cited text no. 112
    
113.
Maier CM, Sun GH, Cheng D, Yenari MA, Chan PH, Steinberg GK. Effects of mild hypothermia on superoxide anion production, superoxide dismutase expression, and activity following transient focal cerebral ischemia. Neurobiol Dis 2002;11:28-42.   Back to cited text no. 113
    
114.
Moro MA, Cárdenas A, Hurtado O, Leza JC, Lizasoain I. Role of nitric oxide after brain ischaemia. Cell Calcium 2004;36:265-75.   Back to cited text no. 114
    
115.
Karabiyikoglu M, Han HS, Yenari MA, Steinberg GK. Attenuation of nitric oxide synthase isoform expression by mild hypothermia after focal cerebral ischemia: Variations depending on timing of cooling. J Neurosurg 2003;98:1271-6.   Back to cited text no. 115
    
116.
Han HS, Qiao Y, Karabiyikoglu M, Giffard RG, Yenari MA. Influence of mild hypothermia on inducible nitric oxide synthase expression and reactive nitrogen production in experimental stroke and inflammation. J Neurosci 2002;22:3921-8.   Back to cited text no. 116
    
117.
Kawabori M, Hokari M, Zheng Z, Kim JY, Calosing C, Hsieh CL, et al. Triggering receptor expressed on myeloid cells-2 correlates to hypothermic neuroprotection in ischemic stroke. Ther Hypothermia Temp Manag 2013;3:189-98.   Back to cited text no. 117
    
118.
Kawabori M, Kacimi R, Kauppinen T, Calosing C, Kim JY, Hsieh CL, et al. Triggering receptor expressed on myeloid cells 2 (TREM2) deficiency attenuates phagocytic activities of microglia and exacerbates ischemic damage in experimental stroke. J Neurosci 2015;35:3384-96.   Back to cited text no. 118
    
119.
Dietrich WD, Busto R, Halley M, Valdes I. The importance of brain temperature in alterations of the blood-brain barrier following cerebral ischemia. J Neuropathol Exp Neurol 1990;49:486-97.   Back to cited text no. 119
    
120.
Kawanishi M, Kawai N, Nakamura T, Luo C, Tamiya T, Nagao S. Effect of delayed mild brain hypothermia on edema formation after intracerebral hemorrhage in rats. J Stroke Cerebrovasc Dis 2008;17:187-95.   Back to cited text no. 120
    
121.
Baumann E, Preston E, Slinn J, Stanimirovic D. Post-ischemic hypothermia attenuates loss of the vascular basement membrane proteins, agrin and SPARC, and the blood-brain barrier disruption after global cerebral ischemia. Brain Res 2009;1269:185-97.   Back to cited text no. 121
    
122.
Candelario-Jalil E, Yang Y, Rosenberg GA. Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience 2009;158:983-94.   Back to cited text no. 122
    
123.
Montaner J, Alvarez-Sabin J, Molina C, Anglés A, Abilleira S, Arenillas J, et al. Matrix metalloproteinase expression after human cardioembolic stroke: Temporal profile and relation to neurological impairment. Stroke 2001;32:1759-66.   Back to cited text no. 123
    
124.
Gidday JM, Gasche YG, Copin JC, Shah AR, Perez RS, Shapiro SD, et al. Leukocyte-derived matrix metalloproteinase-9 mediates blood-brain barrier breakdown and is proinflammatory after transient focal cerebral ischemia. Am J Physiol Heart Circ Physiol 2005;289:H558-68.   Back to cited text no. 124
    
125.
Oda Y, Gao G, Wei EP, Povlishock JT. Combinational therapy using hypothermia and the immunophilin ligand FK506 to target altered pial arteriolar reactivity, axonal damage, and blood-brain barrier dysfunction after traumatic brain injury in rat. J Cereb Blood Flow Metab 2011;31:1143-54.   Back to cited text no. 125
    
126.
Manley GT, Fujimura M, Ma T, Noshita N, Filiz F, Bollen AW, et al. Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med 2000;6:159-63.   Back to cited text no. 126
    
127.
Xiao F, Arnold TC, Zhang S, Brown C, Alexander JS, Carden DL, et al. Cerebral cortical aquaporin-4 expression in brain edema following cardiac arrest in rats. Acad Emerg Med 2004;11:1001-7.   Back to cited text no. 127
    
128.
Tang XN, Liu L, Koike MA, Yenari MA. Mild hypothermia reduces tissue plasminogen activator-related hemorrhage and blood brain barrier disruption after experimental stroke. Ther Hypothermia Temp Manag 2013;3:74-83.   Back to cited text no. 128
    
129.
Fingas M, Penner M, Silasi G, Colbourne F. Treatment of intracerebral hemorrhage in rats with 12 h, 3 days and 6 days of selective brain hypothermia. Exp Neurol 2009;219:156-62.   Back to cited text no. 129
    
130.
Martini WZ. The effects of hypothermia on fibrinogen metabolism and coagulation function in swine. Metabolism 2007;56:214-21.   Back to cited text no. 130
    
131.
Kernie SG, Parent JM. Forebrain neurogenesis after focal Ischemic and traumatic brain injury. Neurobiol Dis 2010;37:267-74.   Back to cited text no. 131
    
132.
Font MA, Arboix A, Krupinski J. Angiogenesis, neurogenesis and neuroplasticity in ischemic stroke. Curr Cardiol Rev 2010;6:238-44.   Back to cited text no. 132
    
133.
Shruster A, Melamed E, Offen D. Neurogenesis in the aged and neurodegenerative brain. Apoptosis 2010;15:1415-21.   Back to cited text no. 133
    
134.
Yenari MA, Han HS. Influence of therapeutic hypothermia on regeneration after cerebral ischemia. Front Neurol Neurosci 2013;32:122-8.  Back to cited text no. 134
    
135.
Saito K, Fukuda N, Matsumoto T, Iribe Y, Tsunemi A, Kazama T, et al. Moderate low temperature preserves the stemness of neural stem cells and suppresses apoptosis of the cells via activation of the cold-inducible RNA binding protein. Brain Res 2010;1358:20-9.   Back to cited text no. 135
    
136.
Kanagawa T, Fukuda H, Tsubouchi H, Komoto Y, Hayashi S, Fukui O, et al. A decrease of cell proliferation by hypothermia in the hippocampus of the neonatal rat. Brain Res 2006;1111:36-40.  Back to cited text no. 136
    
137.
Xiong M, Cheng GQ, Ma SM, Yang Y, Shao XM, Zhou WH. Post-ischemic hypothermia promotes generation of neural cells and reduces apoptosis by Bcl-2 in the striatum of neonatal rat brain. Neurochem Int 2011;58:625-33.   Back to cited text no. 137
    
138.
Silasi G, Colbourne F. Therapeutic hypothermia influences cell genesis and survival in the rat hippocampus following global ischemia. J Cereb Blood Flow Metab 2011;31:1725-35.   Back to cited text no. 138
    
139.
Lasarzik I, Winkelheide U, Thal SC, Benz N, Lörscher M, Jahn-Eimermacher A, et al. Mild hypothermia has no long-term impact on postischemic neurogenesis in rats. Anesth Analg 2009;109:1632-9.   Back to cited text no. 139
    
140.
Lotocki G, de Rivero Vaccari JP, Perez ER, Sanchez-Molano J, Furones-Alonso O, Bramlett HM, et al. Alterations in blood-brain barrier permeability to large and small molecules and leukocyte accumulation after traumatic brain injury: Effects of post-traumatic hypothermia. J Neurotrauma 2009;26:1123-34.   Back to cited text no. 140
    
141.
Bennet L, Roelfsema V, George S, Dean JM, Emerald BS, Gunn AJ. The effect of cerebral hypothermia on white and grey matter injury induced by severe hypoxia in preterm fetal sheep. J Physiol 2007;578:491-506.   Back to cited text no. 141
    
142.
Matijasevic Z, Snyder JE, Ludlum DB. Hypothermia causes a reversible, p53-mediated cell cycle arrest in cultured fibroblasts. Oncol Res 1998;10:605-10.   Back to cited text no. 142
    
143.
Gopurappilly R, Pal R, Mamidi MK, Dey S, Bhonde R, Das AK. Stem cells in stroke repair: Current success and future prospects. CNS Neurol Disord Drug Targets 2011;10:741-56.   Back to cited text no. 143
    
144.
Li L, Harms KM, Ventura PB, Lagace DC, Eisch AJ, Cunningham LA. Focal cerebral ischemia induces a multilineage cytogenic response from adult subventricular zone that is predominantly gliogenic. Glia 2010;58:1610-9.   Back to cited text no. 144
    
145.
Hawthorne AL, Hu H, Kundu B, Steinmetz MP, Wylie CJ, Deneris ES, et al. The unusual response of serotonergic neurons after CNS Injury: Lack of axonal dieback and enhanced sprouting within the inhibitory environment of the glial scar. J Neurosci 2011;31:5605-16.   Back to cited text no. 145
    
146.
Trendelenburg G, Dirnagl U. Neuroprotective role of astrocytes in cerebral ischemia: Focus on ischemic preconditioning. Glia 2005;50:307-20.   Back to cited text no. 146
    
147.
Xie YC, Li CY, Li T, Nie DY, Ye F. Effect of mild hypothermia on angiogenesis in rats with focal cerebral ischemia. Neurosci Lett 2007;422:87-90.  Back to cited text no. 147
    
148.
Kao CH, Chio CC, Lin MT, Yeh CH. Body cooling ameliorating spinal cord injury may be neurogenesis-, anti-inflammation- and angiogenesis-associated in rats. J Trauma 2011;70:885-93.   Back to cited text no. 148
    
149.
Kuo JR, Lo CJ, Chang CP, Lin HJ, Lin MT, Chio CC. Brain cooling-stimulated angiogenesis and neurogenesis attenuated traumatic brain injury in rats. J Trauma 2010;69:1467-72.   Back to cited text no. 149
    
150.
Navarro-Sobrino M, Rosell A, Hernández-Guillamon M, Penalba A, Boada C, Domingues-Montanari S, et al. A large screening of angiogenesis biomarkers and their association with neurological outcome after ischemic stroke. Atherosclerosis 2011;216:205-11.  Back to cited text no. 150
    
151.
Lotocki G, de Rivero Vaccari J, Alonso O, Molano JS, Nixon R, Dietrich WD, et al. Oligodendrocyte vulnerability following traumatic brain injury in rats: Effect of moderate hypothermia. Ther Hypothermia Temp Manag 2011;1:43-51.   Back to cited text no. 151
    
152.
Imada S, Yamamoto M, Tanaka K, Seiwa C, Watanabe K, Kamei Y, et al. Hypothermia-induced increase of oligodendrocyte precursor cells: Possible involvement of plasmalemmal voltage-dependent anion channel 1. J Neurosci Res 2010;88:3457-66.   Back to cited text no. 152
    
153.
Schmitt KR, Boato F, Diestel A, Hechler D, Kruglov A, Berger F, et al. Hypothermia-induced neurite outgrowth is mediated by tumor necrosis factor-alpha. Brain Pathol 2010;20:771-9.  Back to cited text no. 153
    



This article has been cited by
1 Therapeutic hypothermia in patients with poor-grade aneurysmal subarachnoid hemorrhage
So Young Won, Mi Kyung Kim, Jihye Song, Yong Cheol Lim
Clinical Neurology and Neurosurgery. 2022; : 107369
[Pubmed] | [DOI]
2 The blood heat exchanger in intra-arterial selective cooling infusion for acute ischemic stroke: A computational fluid-thermodynamics performance, experimental assessment and evaluation on the brain temperature
Miaowen Jiang, Yuan Gao, Chuanjie Wu, Longfei Wu, Shoucheng Tang, Zhichen Yin, Ang Li, Kun Wang, Shiqiang Zheng, Hangil Lee, Yuchuan Ding, Ming Li, Xunming Ji
Computers in Biology and Medicine. 2022; 145: 105497
[Pubmed] | [DOI]
3 Design and evaluation of an air-insulated catheter for intra-arterial selective cooling infusion from numerical simulation and in vitro experiment
Miaowen Jiang, Ming Li, Yuan Gao, Zhichen Yin, Yuchuan Ding, Yufeng Zheng, Shiqiang Zheng, Chuanjie Wu, Ang Li, Jiancheng Fang, Xunming Ji
Medical Engineering & Physics. 2022; 99: 103736
[Pubmed] | [DOI]
4 Short-duration hypothermia completed prior to reperfusion prevents intracranial pressure elevation following ischaemic stroke in rats
Daniel Omileke, Sara Azarpeykan, Steven W. Bothwell, Debbie Pepperall, Daniel J. Beard, Kirsten Coupland, Adjanie Patabendige, Neil J. Spratt
Scientific Reports. 2021; 11(1)
[Pubmed] | [DOI]
5 Neuroprotective Effects of Early Hypothermia Induced by Phenothiazines and DHC in Ischemic Stroke
Yun Han,Xiao-kun Geng,Hangil Lee,Fengwu Li,Yuchuan Ding,Shao-Hsuan Kao
Evidence-Based Complementary and Alternative Medicine. 2021; 2021: 1
[Pubmed] | [DOI]
6 Hypothermia and Stroke: Pros and Cons
Firoozeh Alavian
The Neuroscience Journal of Shefaye Khatam. 2019; 7(2): 83
[Pubmed] | [DOI]
7 The role of microRNAs in newborn brain development and hypoxic ischaemic encephalopathy
Vennila Ponnusamy,Ping K. Yip
Neuropharmacology. 2019; 149: 55
[Pubmed] | [DOI]
8 Clinical potential of pre-reperfusion hypothermia in ischemic injury
Yun Han,Gary B. Rajah,Mohammed Hussain,Xiaokun Geng
Neurological Research. 2019; : 1
[Pubmed] | [DOI]
9 Brain-selective mild hypothermia promotes long-term white matter integrity after ischemic stroke in mice
Li-Qiang Liu,Xiang-Rong Liu,Jing-Yan Zhao,Feng Yan,Rong-Liang Wang,Shao-Hong Wen,Lei Wang,Yu-Min Luo,Xun-Ming Ji
CNS Neuroscience & Therapeutics. 2018;
[Pubmed] | [DOI]
10 Effects of Therapeutic Hypothermia Combined with Other Neuroprotective Strategies on Ischemic Stroke: Review of Evidence
Zheng Zhang,Linlei Zhang,Yuchuan Ding,Zhao Han,Xunming Ji
Aging and Disease. 2018; 9(3): 507
[Pubmed] | [DOI]
11 Mild Therapeutic Hypothermia Protects the Brain from Ischemia/Reperfusion Injury through Upregulation of iASPP
Xiangrong Liu,Shaohong Wen,Shunying Zhao,Feng Yan,Shangfeng Zhao,Di Wu,Xunming Ji
Aging and Disease. 2018; 9(3): 401
[Pubmed] | [DOI]
12 Synergistically Induced Hypothermia and Enhanced Neuroprotection by Pharmacological and Physical Approaches in Stroke
Jun Zhang,Kaiyin Liu,Omar Elmadhoun,Xunming Ji,Yunxia Duan,Jingfei Shi,Xiaoduo He,Xiangrong Liu,Di Wu,Ruiwen Che,Xiaokun Geng,Yuchuan Ding
Aging and Disease. 2018; 9(4): 578
[Pubmed] | [DOI]
13 Hypoxia, hibernation and Neuroprotection: An Experimental Study in Mice
Changhong Ren,Sijie Li,Gary Rajah,Guo Shao,Guowei Lu,Rongrong Han,Qingjian Huang,Haiyan Li,Yuchuan Ding,Kunlin Jin,Xunming Ji
Aging and Disease. 2018; 9(4): 761
[Pubmed] | [DOI]
14 RNA binding motif protein 3: a potential biomarker in cancer and therapeutic target in neuroprotection
Ren-Bin Zhou,Xiao-Li Lu,Chen-Yan Zhang,Da-Chuan Yin
Oncotarget. 2017; 8(13): 22235
[Pubmed] | [DOI]
15 NOX Activation by Subunit Interaction and Underlying Mechanisms in Disease
Radhika Rastogi,Xiaokun Geng,Fengwu Li,Yuchuan Ding
Frontiers in Cellular Neuroscience. 2017; 10
[Pubmed] | [DOI]
16 Exacerbation of Brain Injury by Post-Stroke Exercise Is Contingent Upon Exercise Initiation Timing
Fengwu Li,Xiaokun Geng,Hajra Khan,John T. Pendy Jr.,Changya Peng,Xiaorong Li,Jose A. Rafols,Yuchuan Ding
Frontiers in Cellular Neuroscience. 2017; 11
[Pubmed] | [DOI]
17 Transcriptome Analysis of Gene Expression Provides New Insights into the Effect of Mild Therapeutic Hypothermia on Primary Human Cortical Astrocytes Cultured under Hypoxia
Mootaz M. Salman,Philip Kitchen,M. Nicola Woodroofe,Roslyn M. Bill,Alex C. Conner,Paul R. Heath,Matthew T. Conner
Frontiers in Cellular Neuroscience. 2017; 11
[Pubmed] | [DOI]
18 Phenothiazines Enhance Mild Hypothermia-induced Neuroprotection via PI3K/Akt Regulation in Experimental Stroke
Hong An,Yunxia Duan,Di Wu,James Yip,Omar Elmadhoun,Joshua C. Wright,Wenjuan Shi,Kaiyin Liu,Xiaoduo He,Jingfei Shi,Fang Jiang,Xunming Ji,Yuchuan Ding
Scientific Reports. 2017; 7(1)
[Pubmed] | [DOI]
19 Relationship between Post-Thrombolysis Blood Pressure and Outcome in Acute Ischemic Stroke Patients Undergoing Thrombolysis Therapy
Longfei Wu,Xiaoqin Huang,Di Wu,Wenbo Zhao,Chuanjie Wu,Ruiwen Che,Zhen Zhang,Fang Jiang,Tingting Bian,Tingting Yang,Kai Dong,Qian Zhang,Zhipeng Yu,Qingfeng Ma,Haiqing Song,Yuchuan Ding,Xunming Ji
Journal of Stroke and Cerebrovascular Diseases. 2017;
[Pubmed] | [DOI]
20 Dihydrocapsaicin (DHC) enhances the hypothermia-induced neuroprotection following ischemic stroke via PI3K/Akt regulation in rat
Di Wu,Jingfei Shi,Omar Elmadhoun,Yunxia Duan,Hong An,Jun Zhang,Xiaoduo He,Ran Meng,Xiangrong Liu,Xunming Ji,Yuchuan Ding
Brain Research. 2017; 1671: 18
[Pubmed] | [DOI]
21 Neuroprotection by Chlorpromazine and Promethazine in Severe Transient and Permanent Ischemic Stroke
Xiaokun Geng,Fengwu Li,James Yip,Changya Peng,Omar Elmadhoun,Jiamei Shen,Xunming Ji,Yuchuan Ding
Molecular Neurobiology. 2016;
[Pubmed] | [DOI]
22 Pharmacological hypothermia: a potential for future stroke therapy?
Kaiyin Liu,Hajra Khan,Xiaokun Geng,Jun Zhang,Yuchuan Ding
Neurological Research. 2016; 38(6): 478
[Pubmed] | [DOI]
23 Local cerebral hypothermia induced by selective infusion of cold lactated ringer’s: a feasibility study in rhesus monkeys
Bincheng Wang,Di Wu,David Dornbos III,Jingfei Shi,Yanhui Ma,Mo Zhang,Yumei Liu,Jian Chen,Yuchuan Ding,Yinghao Luo,Xunming Ji
Neurological Research. 2016; 38(6): 545
[Pubmed] | [DOI]
24 Current AQP research: therapeutic approaches to ischemic and hemorrhagic stroke
Linlin Ma,Longfei Guan,JessieN Ding,Xiaokun Geng
Neural Regeneration Research. 2016; 11(12): 1918
[Pubmed] | [DOI]



 

Top
 
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Preclinical Stud...
Conclusions
References

 Article Access Statistics
    Viewed10695    
    Printed415    
    Emailed0    
    PDF Downloaded968    
    Comments [Add]    
    Cited by others 24    

Recommend this journal