Abstract
Traumatic brain injury (TBI) is a multifaceted disease with intrinsically complex heterogeneity and remains a significant clinical challenge to manage. TBI model systems have demonstrated many mechanisms that contribute to brain parenchymal cell death, including glutamate and calcium toxicity, oxidative stress, inflammation, and mitochondrial dysfunction. Mitochondria are critically regulated by uncoupling proteins (UCP), which allow protons to leak back into the matrix and thus reduce the mitochondrial membrane potential by dissipating the proton motive force. This uncoupling of oxidative phosphorylation from adenosine triphosphate (ATP) synthesis is potentially critical for protection against cellular injury as a result of TBI and stroke. A greater understanding of the underlying mechanism or mechanisms by which uncoupling protein-2 (UCP2) functions to maintain or optimize mitochondrial function, and the conditions which precipitate the failure of these mechanisms, would inform future research and treatment strategies. We posit that UCP2-mediated function underlies the physiological response to neuronal stress associated with traumatic and ischemic injury and that clinical development of UCP2-targeted treatment would significantly impact these patient populations. With a focus on clinical relevance in TBI, we synthesize current knowledge concerning UCP2 and its potential neuroprotective role and apply this body of knowledge to current and potential treatment modalities.
Keywords: Neuronal injury, Mitochondria, Traumatic brain injury, Ischemia, Neuronal cell death
Introduction
Traumatic brain injury (TBI) is a global public health epidemic resulting in over 2.8 million hospitalizations of Americans each year, with more than 500,000 deaths. Because of the composite and destructive effects of both the initial mechanical insult and the secondary injury arising from a variety of biochemical cascades, TBI is a multifaceted disease with intrinsically complex heterogeneity and remains a significant clinical challenge to manage. Experimental TBI models have demonstrated many mechanisms that contribute to neuronal and glial cell death, including glutamate and calcium toxicity, oxidative stress, inflammation, and mitochondrial dysfunction. Mitochondria have been recently established as the principal mediators of cell death, orchestrating the necrotic, apoptotic, and autophagic cellular death pathways [1, 2]. These pathways are critically regulated by uncoupling proteins (UCP), which reduce the mitochondrial membrane potential by causing a proton leak back into the matrix. This process dissipates the proton motive force and uncouples oxidative phosphorylation from adenosine triphosphate (ATP) synthesis [3]. Of the five UCP thus far discovered, emerging evidence identifies uncoupling protein-2 (UCP2) as potentially critical for protection against cellular injury from TBI and stroke [4–8]. With a focus on clinical relevance in TBI, this review article will synthesize the state of our present knowledge concerning UCP2 and its potential neuroprotective role.
Mitochondria
Mitochondrial ATP synthase completes the enzymatic process of oxidative phosphorylation, converting nutrient energy to reduced cofactors (nicotinamide adenine dinucleotide, NADH, and flavin adenine dinucleotide, FADH2) through electron shuttling and a proton motive force (PMF) across the inner mitochondrial membrane. These gradient forces consist of a chemical potential and, more critically, an electrical potential (ΔΨm), which accounts for 90% of the PMF [9–12]. This ΔΨm is the potential energy source that drives phosphorylation of adenosine diphosphate (ADP) to ATP by allowing hydrogen ions to move down their energy gradient through ATP synthase, but is also the source of potentially dangerous reactive oxygen species (ROS) [13, 14]. In the instance of TBI and stroke, ΔΨm is too great, resulting in calcium flows into mitochondria with concurrent production of ROS and subsequent cellular damage, thus initiating the process of cell death. This process occurs through the direct result of the ΔΨm increase triggering the mitochondrial pore transition protein (mPTP) complex [15–18]. An alternative pathway capable of dissipating ΔΨm to reduce mitochondrial calcium load and mPTP activation without increasing oxidative damage is through the function of uncoupling proteins such as UCP2, which account for 95% of all proton movement that bypasses the ATP synthase [19]. These proteins are ubiquitous enough to account for 25% of the standard metabolic rate [20].
UCP2
UCP1, or thermogenin, was discovered in brown adipose tissue where its diversion of metabolic energy to non-shivering thermal energy is recognized for its importance in thermoregulation of newborns as well as in cold-blooded, hibernating, and overfed members of the animal kingdom [21–23]. Discovered in 1997, UCP2 [24] is the best-studied of the non-adipose UCP. UPC2 was well characterized in secondary and tertiary structure in comparison to a bovine ADP/ATP carrier, adenine nucleotide translocase-1 (ANT1), which also permits dissipation of the mitochondrial proton gradient. These two proteins were found to have similar molecular structure despite having only 20% sequence homology [24, 25]. Interestingly, UCP2 appears to have an unusually short half-life of 30 min, compared to its homologue UCP1, which has a half-life of 30 h [26]. This short half-life permits rapid variability of activity at both the mRNA and protein levels [20], suggesting a role in rapid physiological stress responses. UCP2 exists in a glutathione-bound inactive state such that UCP2 activation to reduce ROS production in a stressed cell also liberates a stoichiometric amount of glutathione to mitigate existing ROS moieties [27]. In the central nervous system, mammalian UCP2 mRNA and protein expression is highest in areas that could be described as high-risk for stress. High risk regions would be those with direct access to the blood stream, axons and axon terminals abundant in the hypothalamus, neural circuits important for endocrine and homeostatic regulation [28–30], and those associated with neurodegenerative disease such as the substantia nigra and locus coeruleus [29–32]. UCP2 is recognized as a factor responsible for increased neuronal survival in stressed states such as after stroke or trauma [33–35] through several mechanisms which coalesce at the dissipation and subsequent stabilization of mitochondrial ΔΨm [31, 32, 36–39], as has been confirmed in transfected cell culture [39].
Physiology of UCP2
Oxidative phosphorylation
UCP dissociate oxidation from phosphorylation, suppressing ATP production per mitochondrion under physiological conditions. During normal cellular function this bypassing of ATP synthase would result in a dangerous and wasteful futile cycle, but under conditions in which ATP production is limited due to lack of necessary substrates, including oxygen, the dissipation of ΔΨm through other means would be necessary to protect against ROS damage and initiation of apoptosis [19, 40, 41]. Experimental data suggest that UCP2 contributes to neuronal stress tolerance in acute settings, which over time triggers mitochondrial proliferation and increased ATP/ADP production [32, 42]. As ΔΨm is intimately linked to ATP production, a disruption of delivery or ability to effectively utilize substrates for oxidative phosphorylation would lead to ΔΨm increasing to dangerous levels with subsequent ROS production, calcium influx into mitochondria and eventually cell death [39, 40, 43–45]. Recent evidence further indicates that the principle function of UCP2 is to dissipate ΔΨm in vivo [46], which may underlie the emerging neuroprotective role of UCP2 in TBI.
Calcium regulation and apoptosis
Mitochondria represent a point of integration for cell death signaling in vertebrates through the activation of mitochondrial membrane permeabilization [1, 40, 44, 47, 48], releasing myriad cell death factors associated with apoptosis, necrosis, or the recently described hybrid necroptosis [45], and host both pro-survival and pro-apoptotic factors critical for regulating mitochondrial membrane permeability [49]. Regulation of calcium, particularly the shifting of calcium load into mitochondria, is intimately linked to both the necrotic and apoptotic cell death pathways [45, 50]. UCP2 has been identified as a regulator of calcium homeostasis, whose role is best appreciated in cases of cellular stress [51, 52]. Calcium influx into mitochondria increases with a greater ΔΨm, as does ROS production [19, 41]. Thus UCP2 overexpression resulted in decreased ΔΨm restricting calcium influx into mitochondria and decreased ROS production [39, 43]. The activation of and interaction between several mitochondrial channels, including calcium channels, activated directly or indirectly through mitochondrial ΔΨm, changes under the control of UCP2 and emphasizes the importance of the uncoupler [40, 44, 48].
Reactive oxygen species (ROS)
Reactive oxygen species (ROS), both radicals and non-radicals (hydrogen peroxide), are highly reactive byproducts of oxidative respiration present in all cells that function as signaling elements at physiological concentrations [19, 53]. Mitochondrial ROS production is highly sensitive to slight changes in ΔΨm [54–56]. UCP2 lowers ΔΨm with the subsequent decrease of ROS production [38, 42, 57, 58]; conversely, in UCP2 knockout animals, an increase in ΔΨm is observed, which promotes ROS production [42, 59]. Interestingly, UCP2 also serves as a cellular target of ROS signals such as superoxide, which activates UCP2 to reduce ΔΨm and lower ROS generation [60]. Recent evidence further supports the notion that rising levels of ROS induce mitochondrial uncoupling through UCP2 activation, a process which also liberates glutathione [27], indicating that under physiological conditions UCP2 is part of an adaptive response to maintain proper redox homeostasis through bidirectional regulation.
Physiological compensation
The physiological role of UCP2 is thus one of compensations to decrease ΔΨm in the face of cellular stress. UCP2 is central to the diabetes-related stress response of pancreatic islet cells and has been used as a marker of sublethal insult to beta cells [61, 62], eventually resulting in activation of the mPTP [63]. This shift from effective compensation to mPTP activation and decompensation may underlie recent observations that the peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist rosiglitazone, known to cause oxidative damage to cardiomyocytes, has an acute protective effect in cultured cells [14] and neurons [64]. In the case of TBI, the role of UCP2 would be of acute compensation, potentially mitigating damage enough to allow cellular recovery in cases where UCP2 is not overwhelmed. Exemplifying this mechanism is ghrelin, which has been shown to be neuroprotective in the context of TBI through a mechanism reminiscent of ischemic preconditioning, resulting in increased UCP2 expression and greater neuroprotection [7, 8, 65, 66]. In contrast, ghrelin’s counterpart leptin has been shown to have more acute UCP2-dependent mitochondrial stabilization effects in thalamic neurons [64, 67]. It appears that in neurons, UCP2 stabilizes mitochondria by responding to sublethal stress.
Pathophysiology of UCP2
Neurodegeneration
ROS, particularly hydrogen peroxide, have come to be appreciated as important for cellular “redox” signaling to mediate diverse physiological responses [36, 68–70], notably synaptic signaling and plasticity [71], where tight spatial and temporal regulations are likely critical to the ability of ROS moieties to perform physiologic functions with minimal toxicity [72]. The importance of ROS-mediated neurotoxicity in ischemic and traumatic brain injury as well as in chronic neurodegenerative diseases has been well documented, and the presence of free radicals in the brain resulting from mitochondrial dysfunction or excitotoxicity has been shown to contribute to neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis [73]. Damage to respiratory chain Complex IV has been shown to contribute to multiple sclerosis pathophysiology by increasing ΔΨm and augmenting glutamate-mediated axonal injury and associated immune activity and demyelination [74], while robustly functioning mitochondria are associated with faster and more complete recovery [75].
Amyloid-β is recognized as a part of Alzheimer’s disease pathophysiology. While neuroprotective in the initial phase [76], its accumulation is damaging and not unique to Alzheimer’s disease alone: amyloid-β has also been found to accumulate in TBI patients [77–79]. Oxidative stress in the context of TBI was demonstrated to increase production of amyloid in the brain. In controlled studies of TBI, it was found that levels of ROS were elevated within one hour after injury and persisted at elevated levels for more than 4 days [80].
A post-mortem human study of neurons from the substantia nigra found that idiopathic Parkinson’s disease demonstrated increased respiratory chain complexes in mitochondria with the same density of mitochondria [81], possibly representing mitochondrial compensation in response to the suboptimal efficacy of the neuronal system. One recent study identified ursocholanic acid as a compound which acted to recover mitochondrial function and normalize ATP levels in fibroblasts from patients with a parkin (PARK2) mutation [82]. Importantly, the cell donors were patients with a predisposition to a Parkinson diagnosis, not those with diagnosed Parkinson’s disease. The most attractive feature of such approaches to finding novel treatments of neurodegenerative disease is the recognition that the optimal time to treat is during the subclinical compensatory phase, before failure of the system allows diagnosis.
Reperfusion injury and UCP2
Post-traumatic oxygen delivery is an important part of emergent therapy to prevent complications of TBI, such that hyperbaric oxygen has been studied alongside high-flow nasal cannula to optimize oxygen delivery to brain parenchyma without inducing oxygen toxicity. Both 60-min bursts of hyperbaric oxygen and three-hour treatments with high-flow nasal cannula were shown to be efficacious, increasing cerebral blood flow and decreasing lactate levels with no sign of pulmonary or cerebral damage in patients with severe TBI [83]. Murine hyperoxia has been shown to induce UCP2 within 60 min, perhaps in response to the sudden increase in ROS production, suggesting a mechanism for this protection in which UCP2 was not being transcribed anew but that existing UCP2 mRNA and glutathione-coupled UCP2 were being mobilized; indeed, UCP2 protein level was found to increase with a corresponding decrease in ΔΨm while the amount of UCP2 mRNA did not change [84]. This result extends specifically to toxicity mediated by hyperactivity associated with seizure in a murine model and is postulated to be at the core of the preconditioning phenomenon of cerebral vascular injury protection [32]. Oxygen delivery is universally recognized as critically important to the preservation of function in cerebral injury, but is also perhaps only important given the assumed preservation of the ability of cerebral parenchyma to utilize the oxygen with functioning mitochondria [85].
We hypothesize that UCP2 is of universal importance for helping cells overcome periods of stress. UCP2 is also part of the decision-making machinery that pivots the cell from a defensive posture of compensation to an apoptotic state upon decompensation. Through its capacities to regulate cellular energy balance and metabolism, calcium homeostasis, and ROS production, UCP2 plays a critical role in deciding the fate of the cell through mitochondrial membrane permeabilization [4]. For example, the principal trigger for mPTP opening is intramitochondrial calcium in the presence of phosphate – a state consistent with a large ΔΨm and inefficiently functioning ATP-synthase, which favors ROS production [44, 86]. UCP2 may inhibit the activation of mPTP by mildly depolarizing mitochondria, leading to a decreased uptake of calcium, a reduced production of ROS, and an altered ATP/ADP ratio during compensation [4]. However, this compensation may be overcome at a critical point leading to triggering of mPTP and subsequent cell death. Understanding the underlying mechanism of cellular deterioration may allow new and more effective treatment strategies [87], such as understanding the metabolically distinct state of stressed cells. In this situation, even the biosynthetic pentose phosphate shunt can become clinically toxic [88], as the restoration of normal levels of oxygen and glucose exceeds the limits of this abnormal physiological system and leads to reperfusion injury.
UCP2 induction has been found to be protective against ischemia-reperfusion injury and may be a common downstream factor in multiple pathways including ischemic preconditioning protection in human neurons [64]. In contrast to UCP1, UCP2 does not contribute to core body thermogenesis; however, intrinsic to its ability to uncouple oxidative phosphorylation, UCP2 dissipates energy in the form of heat at the mitochondrial level. Because neurotransmission is highly temperature sensitive [89, 90] and neuronal UCP2-containing mitochondria appear to accumulate in axon terminals [29], experimental data suggest that UCP2 may function to generate a synaptic microenvironmental temperature gradient and thermally modulate neurotransmission [29, 91]. The temperature-dependent nature of neurotransmitter diffusion and receptor kinetics has been well established [92, 93]. Recent evidence further supports that both synaptic and volume diffusion of neurotransmitters are highly temperature sensitive [89, 90]. Further, neurotransmission is highly sensitive to cellular mechanisms that regulate calcium and ROS homeostasis [50, 71, 94].
Traumatic brain injury (TBI)
The effects of TBI on brain metabolism may also have a central role, and the neuroprotection offered by therapeutic hypothermia has been linked to a reduced blood glucose and lactate level as well as antioxidant preservation [95, 96]; all metabolic effects associated with preservation of mitochondrial functionality. Much more is known about ice-bath induced and cold saline infusion-mediated therapeutic hypothermia, and as the lack of harm associated with pre-hospital cooling has recently been demonstrated [97], more clinical studies are likely to come. Interestingly, a high blood alcohol level at the time of TBI has been associated with improved outcome in therapeutic hypothermia, leading to hypotheses about the optimal composition of intravascular contents during therapeutic hypothermia. One recent study, which demonstrated such a superior outcome for patients that were considered alcohol positive (alcohol ≥ 0.08 g/dL, i.e. ≥ 17.4 mmol/L) at the time of admission, opens the possibility that these patients may have been prone to an undeserved ‘severe’ subjective injury rating due to cognitive effects of inebriation, possibly explaining why severe TBI patients admitted with high blood alcohol levels recovered to a greater extent than their sober counterparts [98]. We propose that the underlying cellular mechanisms of therapeutic hypothermia be probed in order to identify more clearly how reperfusion injury is mitigated, and we posit that UCP2 may be central both to cellular protection from ROS and to the signaling pathway resulting in decompensation at the cellular level.
Diffuse axonal injury as a consequence of TBI includes direct axonal injury and myelin disruption and leads to major impairment of many functional pathways [99]. With the disruption of the integration necessary for cognitive and executive function, patients may be significantly impaired, though new rehabilitative techniques are emerging that can help to improve higher-order functioning at least in mild TBI cases [100]. Gaining an understanding of the cellular processes which govern the response to and control of damage, such as those involved in apoptotic signaling or reducing ROS exposure, would offer a method of reducing axonal injury or enhancing recovery in the early post-traumatic phase.
Hypothermia seeks to decrease ROS production by decreasing energy demand, but has significant issues in terms of methods applied to achieve cooling [101, 102]. With a greater understanding of the metabolic pathways responsible for the reduced ROS-associated cellular damage as a result of cooling, one could pharmacologically target those pathways and allow rapid and effective pre-hospital or in-hospital cooling effects without dependence on ambulance travel times or comorbidities such as chronic heart failure that may preclude some cooling strategies [101]. Given that UCP2 would dissipate ΔΨm through heat generation and release, some manner of cooling would likely remain necessary but it is possible that the target temperatures may be significantly closer to normal body temperature given the targeted nature of such a therapy. The key to the attractiveness of a UCP2 activation approach to cooling is that as excess ΔΨm dissipates and the danger of ROS-mediated damage is reduced, the substrate for UCP2-generated heat would also be reduced in real time, offering not only a tunable property in ΔΨm response to such a therapy but also a prognostic indicator in the form of clinically measureable temperature changes.
Conclusion
Early intervention to prevent initial neuronal injury extension after TBI is critical for TBI recovery, and UCP2 may offer an important target of early treatment. The efficient delivery of oxygen and glucose to neural tissues after injury has been emphasized [83, 103], but the usefulness of these depends upon the retention of cellular ability to process and utilize them [85]. Post-injury mitochondrial uncoupling can increase tissue salvaged and cognition retained in model systems [104] and may be at least as important as maintaining oxygenation in ischemic neural tissue [85]. While the pleotropic effects of hypothermia have been shown to be efficacious [105] and may include the preservation of mitochondrial function [106], the targeting of UCP2 may provide a specific and reliable treatment for TBI patients to minimize injury and maximize recovery. New techniques are emerging at both the subcellular level [53] and the tissue level [107] to understand the processes underlying TBI, and the development of new techniques to more specifically prevent damage to brain parenchyma will undoubtedly result in improved patient outcomes.
Acknowledgment
Conflict of interest statement: The authors declare that there are no conflicts of interest.
References
- 1.Nagley P, Higgins GC, Atkin JD, Beart PM. Multifaceted deaths orchestrated by mitochondria in neurones. Biochim Biophys Acta. 2010;1802:167–185. doi: 10.1016/j.bbadis.2009.09.004. [DOI] [PubMed] [Google Scholar]
- 2.Schon EA, Przedborski S. Mitochondria: the next (neurode) generation. Neuron. 2011;70:1033–1053. doi: 10.1016/j.neuron.2011.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Krauss S, Zhang CY, Lowell BB. The mitochondrial uncoupling-protein homologues. Nat Rev Mol Cell Biol. 2005;6:248–261. doi: 10.1038/nrm1592. [DOI] [PubMed] [Google Scholar]
- 4.Mattiasson G, Shamloo M, Gido G, Mathi K, Tomasevic G, Yi S, et al. Uncoupling protein-2 prevents neuronal death and diminishes brain dysfunction after stroke and brain trauma. Nat Med. 2003;9:1062–1068. doi: 10.1038/nm903. [DOI] [PubMed] [Google Scholar]
- 5.Mattiasson G, Sullivan PG. The emerging functions of UCP2 in health, disease, and therapeutics. Antioxid Redox Signal. 2006;8:1–38. doi: 10.1089/ars.2006.8.1. [DOI] [PubMed] [Google Scholar]
- 6.Paradis E, Clavel S, Bouillaud F, Ricquier D, Richard D. Uncoupling protein 2: a novel player in neuroprotection. Trends Mol Med. 2003;9:522–525. doi: 10.1016/j.molmed.2003.10.009. [DOI] [PubMed] [Google Scholar]
- 7.Liu Y, Chen L, Xu X, Vicaut E, Sercombe R. Both ischemic preconditioning and ghrelin administration protect hippocampus from ischemia/reperfusion and upregulate uncoupling protein-2. BMC Physiol. 2009;9:17. doi: 10.1186/1472-6793-9-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lopez NE, Gaston L, Lopez KR, Coimbra RC, Hageny A, Putnam J, et al. Early ghrelin treatment attenuates disruption of the blood brain barrier and apoptosis after traumatic brain injury through a UCP-2 mechanism. Brain Res. 2012;1489:140–148. doi: 10.1016/j.brainres.2012.10.031. [DOI] [PubMed] [Google Scholar]
- 9.Klingenberg M. Wanderings in bioenergetics and biomembranes. Biochim Biophys Acta. 2010;1797:579–594. doi: 10.1016/j.bbabio.2010.02.012. [DOI] [PubMed] [Google Scholar]
- 10.Hosler JP, Ferguson-Miller S, Mills DA. Energy transduction: proton transfer through the respiratory complexes. Ann Rev Biochem. 2006;75:165–187. doi: 10.1146/annurev.biochem.75.062003.101730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dzbek J, Korzeniewski B. Control over the contribution of the mitochondrial membrane potential (ΔΨ) and proton gradient (ΔpH) to the protonmotive force (Δp). In silico studies. J Biol Chem. 2008;283:33232–33239. doi: 10.1074/jbc.M802404200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Votyakova TV, Reynolds IJ. ΔΨm-Dependent and -independent production of reactive oxygen species by rat brain mitochondria. J Neurochem. 2001;79:266–277. doi: 10.1046/j.1471-4159.2001.00548.x. [DOI] [PubMed] [Google Scholar]
- 13.Andreyev AY, Kushnareva YE, Starkov AA. Mitochondrial metabolism of reactive oxygen species. Biochemistry (Moscow) 2005;70:200–214. doi: 10.1007/s10541-005-0102-7. [DOI] [PubMed] [Google Scholar]
- 14.Olsen RK, Cornelius N, Gregersen N. Genetic and cellular modifiers of oxidative stress: what can we learn from fatty acid oxidation defects? Mol Genet Metab. 2013;110(Suppl):S31–39. doi: 10.1016/j.ymgme.2013.10.007. [DOI] [PubMed] [Google Scholar]
- 15.Lifshitz J, Sullivan PG, Hovda DA, Wieloch T, McIntosh TK. Mitochondrial damage and dysfunction in traumatic brain injury. Mitochondrion. 2004;4:705–713. doi: 10.1016/j.mito.2004.07.021. [DOI] [PubMed] [Google Scholar]
- 16.Ankarcrona M, Dypbukt JM, Bonfoco E, Zhivotovsky B, Orrenius S, Lipton SA, et al. Glutamate-induced neuronal death: a succession of necrosis or apoptosis depending on mitochondrial function. Neuron. 1995;15:961–973. doi: 10.1016/0896-6273(95)90186-8. [DOI] [PubMed] [Google Scholar]
- 17.Okonkwo DO, Povlishock JT. An intrathecal bolus of cyclosporin A before injury preserves mitochondrial integrity and attenuates axonal disruption in traumatic brain injury. J Cereb Blood Flow Metab. 1999;19:443–451. doi: 10.1097/00004647-199904000-00010. [DOI] [PubMed] [Google Scholar]
- 18.Fiskum G. Mitochondrial participation in ischemic and traumatic neural cell death. J Neurotrauma. 2000;17:843–855. doi: 10.1089/neu.2000.17.843. [DOI] [PubMed] [Google Scholar]
- 19.Brand MD, Affourtit C, Esteves TC, Green K, Lambert AJ, Miwa S, et al. Mitochondrial superoxide: production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med. 2004;37:755–767. doi: 10.1016/j.freeradbiomed.2004.05.034. [DOI] [PubMed] [Google Scholar]
- 20.Azzu V, Brand MD. The on-off switches of the mitochondrial uncoupling proteins. Trends Biochem Sci. 2010;35:298–307. doi: 10.1016/j.tibs.2009.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84:277–359. doi: 10.1152/physrev.00015.2003. [DOI] [PubMed] [Google Scholar]
- 22.Richard D, Picard F. Brown fat biology and thermogenesis. Front Biosci. 2011;16:1233–1260. doi: 10.2741/3786. [DOI] [PubMed] [Google Scholar]
- 23.Nicholls DG, Locke RM. Thermogenic mechanisms in brown fat. Physiol Rev. 1984;64:1–64. doi: 10.1152/physrev.1984.64.1.1. [DOI] [PubMed] [Google Scholar]
- 24.Ricquier D, Bouillaud F. The uncoupling protein homologues: UCP1 UCP2 UCP3, StUCP and AtUCP. Biochem J. 2000;345:161–179. [PMC free article] [PubMed] [Google Scholar]
- 25.Berardi MJ, Shih WM, Harrison SC, Chou JJ. Mitochondrial uncoupling protein 2 structure determined by NMR molecular fragment searching. Nature. 2011;476:109–113. doi: 10.1038/nature10257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Rousset S, Mozo J, Dujardin G, Emre Y, Masscheleyn S, Ricquier D, et al. UCP2 is a mitochondrial transporter with an unusual very short half-life. FEBS Lett. 2007;581:479–482. doi: 10.1016/j.febslet.2007.01.010. [DOI] [PubMed] [Google Scholar]
- 27.Mailloux RJ, Seifert EL, Bouillaud F, Aguer C, Collins S, Harper ME. Glutathionylation acts as a control switch for uncoupling proteins UCP2 and UCP3. J Biol Chem. 2011;286:21865–21875. doi: 10.1074/jbc.M111.240242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Diano S, Urbanski HF, Horvath B, Bechmann I, Kagiya A, Nemeth G, et al. Mitochondrial uncoupling protein 2 (UCP2) in the nonhuman primate brain and pituitary. Endocrinology. 2000;141:4226–4238. doi: 10.1210/endo.141.11.7740. [DOI] [PubMed] [Google Scholar]
- 29.Horvath TL, Warden CH, Hajos M, Lombardi A, Goglia F, Diano S. Brain uncoupling protein 2: uncoupled neuronal mitochondria predict thermal synapses in homeostatic centers. J Neurosci. 1999;19:10417–10427. doi: 10.1523/JNEUROSCI.19-23-10417.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Richard D, Rivest R, Huang Q, Bouillaud F, Sanchis D, Champigny O, et al. Distribution of the uncoupling protein 2 mRNA in the mouse brain. J Comp Neurol. 1998;397:549–560. [PubMed] [Google Scholar]
- 31.Horvath TL, Diano S, Leranth C, Garcia-Segura LM, Cowley MA, Shanabrough M, et al. Coenzyme Q induces nigral mitochondrial uncoupling and prevents dopamine cell loss in a primate model of Parkinson’s disease. Endocrinology. 2003;144:2757–2760. doi: 10.1210/en.2003-0163. [DOI] [PubMed] [Google Scholar]
- 32.Diano S, Matthews RT, Patrylo P, Yang L, Beal MF, Barnstable CJ, et al. Uncoupling protein 2 prevents neuronal death including that occurring during seizures: a mechanism for preconditioning. Endocrinology. 2003;144:5014–5021. doi: 10.1210/en.2003-0667. [DOI] [PubMed] [Google Scholar]
- 33.Mehta SL, Li PA. Neuroprotective role of mitochondrial uncoupling protein 2 in cerebral stroke. J Cereb Blood Flow Metab. 2009;29:1069–1078. doi: 10.1038/jcbfm.2009.4. [DOI] [PubMed] [Google Scholar]
- 34.Slemmer JE, Zhu C, Landshamer S, Trabold R, Grohm J, Ardeshiri A, et al. Causal role of apoptosis-inducing factor for neuronal cell death following traumatic brain injury. Am J Pathol. 2008;173:1795–1805. doi: 10.2353/ajpath.2008.080168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bechmann I, Diano S, Warden CH, Bartfai T, Nitsch R, Horvath TL. Brain mitochondrial uncoupling protein 2 (UCP2): a protective stress signal in neuronal injury. Biochem Pharmacol. 2002;64:363–367. doi: 10.1016/s0006-2952(02)01166-8. [DOI] [PubMed] [Google Scholar]
- 36.Rhee SG. Cell signaling. H2O2, a necessary evil for cell signaling. Science. 2006;312:1882–1883. doi: 10.1126/science.1130481. [DOI] [PubMed] [Google Scholar]
- 37.Negre-Salvayre A, Hirtz C, Carrera G, Cazenave R, Troly M, Salvayre R, et al. A role for uncoupling protein-2 as a regulator of mitochondrial hydrogen peroxide generation. FASEB J. 1997;11:809–815. [PubMed] [Google Scholar]
- 38.Sullivan PG, Dube C, Dorenbos K, Steward O, Baram TZ. Mitochondrial uncoupling protein-2 protects the immature brain from excitotoxic neuronal death. Ann Neurol. 2003;53:711–717. doi: 10.1002/ana.10543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yamada S, Isojima Y, Yamatodani A, Nagai K. Uncoupling protein 2 influences dopamine secretion in PC12h cells. J Neurochem. 2003;87:461–469. doi: 10.1046/j.1471-4159.2003.02005.x. [DOI] [PubMed] [Google Scholar]
- 40.Dejean LM, Martinez-Caballero S, Kinnally KW. Is MAC the knife that cuts cytochrome c from mitochondria during apoptosis? Cell Death Differ. 2006;13:1387–1395. doi: 10.1038/sj.cdd.4401949. [DOI] [PubMed] [Google Scholar]
- 41.Nicholls DG, Ward MW. Mitochondrial membrane potential and neuronal glutamate excitotoxicity: mortality and millivolts. Trends Neurosci. 2000;23:166–174. doi: 10.1016/s0166-2236(99)01534-9. [DOI] [PubMed] [Google Scholar]
- 42.Andrews ZB, Horvath B, Barnstable CJ, Elsworth J, Yang L, Beal MF, et al. Uncoupling protein-2 is critical for nigral dopamine cell survival in a mouse model of Parkinson’s disease. J Neurosci. 2005;25:184–191. doi: 10.1523/JNEUROSCI.4269-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Teshima Y, Akao M, Jones SP, Marban E. Uncoupling protein-2 overexpression inhibits mitochondrial death pathway in cardiomyocytes. Circ Res. 2003;93:192–200. doi: 10.1161/01.RES.0000085581.60197.4D. [DOI] [PubMed] [Google Scholar]
- 44.Kinnally KW, Peixoto PM, Ryu SY, Dejean LM. Is mPTP the gatekeeper for necrosis, apoptosis, or both? Biochim Biophys Acta. 2011;1813:616–622. doi: 10.1016/j.bbamcr.2010.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16:3–11. doi: 10.1038/cdd.2008.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Park D, Han CZ, Elliott MR, Kinchen JM, Trampont PC, Das S, et al. Continued clearance of apoptotic cells critically depends on the phagocyte Ucp2 protein. Nature. 2011;477:220–224. doi: 10.1038/nature10340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kinnally KW, Martinez-Caballero S, Dejean LM. Detection of the mitochondrial apoptosis-induced channel (MAC) and its regulation by Bcl-2 family proteins. Curr Protoc Toxicol. 2006;2:12. doi: 10.1002/0471140856.tx0212s30. [DOI] [PubMed] [Google Scholar]
- 48.Shoshan-Barmatz V, De Pinto V, Zweckstetter M, Raviv Z, Keinan N, Arbel N. VDAC, a multi-functional mitochondrial protein regulating cell life and death. Mol Aspects Med. 2010;31:227–285. doi: 10.1016/j.mam.2010.03.002. [DOI] [PubMed] [Google Scholar]
- 49.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev. 2007;87:99–163. doi: 10.1152/physrev.00013.2006. [DOI] [PubMed] [Google Scholar]
- 50.Berridge MJ, Bootman MD, Lipp P. Calcium - a life and death signal. Nature. 1998;395:645–648. doi: 10.1038/27094. [DOI] [PubMed] [Google Scholar]
- 51.Trenker M, Malli R, Fertschai I, Levak-Frank S, Graier WF. Uncoupling proteins 2 and 3 are fundamental for mitochondrial Ca2+ uniport. Nat Cell Biol. 2007;9:445–452. doi: 10.1038/ncb1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Robinson AD, Ramanathan KB, McGee JE, Newman KP, Weber KT. Oxidative stress and cardiomyocyte necrosis with elevated serum troponins: pathophysiologic mechanisms. Am J Med Sci. 2011;342:129–134. doi: 10.1097/MAJ.0b013e3182231ee3. [DOI] [PubMed] [Google Scholar]
- 53.Niethammer P, Kueh HY, Mitchison TJ. Spatial patterning of metabolism by mitochondria, oxygen, and energy sinks in a model cytoplasm. Curr Biol. 2008;18:586–591. doi: 10.1016/j.cub.2008.03.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang W, Fang H, Groom L, Cheng A, Zhang W, Liu J, et al. Superoxide flashes in single mitochondria. Cell. 2008;134:279–290. doi: 10.1016/j.cell.2008.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pouvreau S. Superoxide flashes in mouse skeletal muscle are produced by discrete arrays of active mitochondria operating coherently. PLoS One. 2010;5:e13035. doi: 10.1371/journal.pone.0013035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Aon MA, Cortassa S, Marban E, O’Rourke B. Synchronized whole cell oscillations in mitochondrial metabolism triggered by a local release of reactive oxygen species in cardiac myocytes. J Biol Chem. 2003;278:44735–44744. doi: 10.1074/jbc.M302673200. [DOI] [PubMed] [Google Scholar]
- 57.Skulachev VP. Role of uncoupled and non-coupled oxidations in maintenance of safely low levels of oxygen and its one-electron reductants. Q Rev Biophys. 1996;29:169–202. doi: 10.1017/s0033583500005795. [DOI] [PubMed] [Google Scholar]
- 58.Skulachev VP. Uncoupling: new approaches to an old problem of bioenergetics. Biochim Biophys Acta. 1998;1363:100–124. doi: 10.1016/s0005-2728(97)00091-1. [DOI] [PubMed] [Google Scholar]
- 59.Arsenijevic D, Onuma H, Pecqueur C, Raimbault S, Manning BS, Miroux B, et al. Disruption of the uncoupling protein-2 gene in mice reveals a role in immunity and reactive oxygen species production. Nat Genet. 2000;26:435–439. doi: 10.1038/82565. [DOI] [PubMed] [Google Scholar]
- 60.Echtay KS, Roussel D, St-Pierre J, Jekabsons MB, Cadenas S, Stuart JA, et al. Superoxide activates mitochondrial uncoupling proteins. Nature. 2002;415:96–99. doi: 10.1038/415096a. [DOI] [PubMed] [Google Scholar]
- 61.Li N, Stojanovski S, Maechler P. Mitochondrial hormesis in pancreatic beta cells: does uncoupling protein 2 play a role? Oxid Med Cell Longev. 2012:740849. doi: 10.1155/2012/740849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chan CB, De Leo D, Joseph JW, McQuaid TS, Ha XF, Xu F, et al. Increased uncoupling protein-2 levels in beta-cells are associated with impaired glucose-stimulated insulin secretion: mechanism of action. Diabetes. 2001;50:1302–1310. doi: 10.2337/diabetes.50.6.1302. [DOI] [PubMed] [Google Scholar]
- 63.Graw JA, von Haefen C, Poyraz D, Möbius N, Sifringer M, Spies CD. Chronic alcohol consumption increases the expression of uncoupling protein-2 and -4 in the brain. Alcohol Clin Exp Res. 2013;37:1650–1656. doi: 10.1111/acer.12144. [DOI] [PubMed] [Google Scholar]
- 64.Kleinridders A, Lauritzen HP, Ussar S, Christensen JH, Mori MA, Bross P, et al. Leptin regulation of Hsp60 impacts hypothalamic insulin signaling. J Clin Invest. 2013;123:4667–4680. doi: 10.1172/JCI67615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lopez NE, Krzyzaniak MJ, Blow C, Putnam J, Ortiz-Pomales Y, Hageny AM, et al. Ghrelin prevents disruption of the blood-brain barrier after traumatic brain injury. J Neurotrauma. 2012;29:385–393. doi: 10.1089/neu.2011.2053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Qi L, Cui X, Dong W, Barrera R, Nicastro J, Coppa GF, et al. Ghrelin attenuates brain injury after traumatic brain injury and uncontrolled hemorrhagic shock in rats. Mol Med. 2012;18:186–193. doi: 10.2119/molmed.2011.00390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Henry BA, Andrews ZB, Rao A, Clarke IJ. Central leptin activates mitochondrial function and increases heat production in skeletal muscle. Endocrinology. 2011;152:2609–2618. doi: 10.1210/en.2011-0143. [DOI] [PubMed] [Google Scholar]
- 68.Stone JR, Yang S. Hydrogen peroxide: a signaling messenger. Antioxid Redox Signal. 2006;8:243–270. doi: 10.1089/ars.2006.8.243. [DOI] [PubMed] [Google Scholar]
- 69.Veal EA, Day AM, Morgan BA. Hydrogen peroxide sensing and signaling. Mol Cell. 2007;26:1–14. doi: 10.1016/j.molcel.2007.03.016. [DOI] [PubMed] [Google Scholar]
- 70.Huang YZ, McNamara JO. Neuroprotective effects of reactive oxygen species mediated by BDNF-independent activation of TrkB. J Neurosci. 2012;32:15521–15532. doi: 10.1523/JNEUROSCI.0755-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Massaad CA, Klann E. Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid Redox Signal. 2011;14:2013–2054. doi: 10.1089/ars.2010.3208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Veal E, Day A. Hydrogen peroxide as a signaling molecule. Antioxid Redox Signal. 2011;15:147–151. doi: 10.1089/ars.2011.3968. [DOI] [PubMed] [Google Scholar]
- 73.Uttara B, Singh AV, Zamboni P, Mahajan RT. Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol. 2009;7:65–74. doi: 10.2174/157015909787602823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mahad DJ, Ziabreva I, Campbell G, Lax N, White K, Hanson PS, et al. Mitochondrial changes within axons in multiple sclerosis. Brain. 2009;132:1161–1174. doi: 10.1093/brain/awp046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ciccarelli O, Altmann DR, McLean MA, Wheeler-Kingshott CA, Wimpey K, Miller DH, et al. Spinal cord repair in MS: does mitochondrial metabolism play a role? Neurology. 2010;74:721–727. doi: 10.1212/WNL.0b013e3181d26968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ayton S, Zhang M, Roberts BR, Lam LQ, Lind M, McLean C, et al. Ceruloplasmin and β-amyloid precursor protein confer neuroprotection in traumatic brain injury and lower neuronal iron. Free Radic Biol Med. 2014;69:331–337. doi: 10.1016/j.freeradbiomed.2014.01.041. [DOI] [PubMed] [Google Scholar]
- 77.Gatson JW, Warren V, Abdelfattah K, Wolf S, Hynan LS, Moore C, et al. Detection of β-amyloid oligomers as a predictor of neurological outcome after brain injury. J Neurosurg. 2013;118:1336–1342. doi: 10.3171/2013.2.JNS121771. [DOI] [PubMed] [Google Scholar]
- 78.Marklund N, Farrokhnia N, Hånell A, Vanmechelen E, Enblad P, Zetterberg H, et al. Monitoring of β-amyloid dynamics after human traumatic brain injury. J Neurotrauma. 2013;31:42–55. doi: 10.1089/neu.2013.2964. [DOI] [PubMed] [Google Scholar]
- 79.Tsitsopoulos PP, Marklund N. Amyloid-β peptides and tau protein as biomarkers in cerebrospinal and interstitial fluid following traumatic brain injury: a review of experimental and clinical studies. Front Neurol. 2013;4:79. doi: 10.3389/fneur.2013.00079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhang QG, Laird MD, Han D, Nguyen K, Scott E, Dong Y, et al. Critical role of NADPH oxidase in neuronal oxidative damage and microglia activation following traumatic brain injury. PLoS One. 2012;7:e34504. doi: 10.1371/journal.pone.0034504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Reeve AK, Park TK, Jaros E, Campbell GR, Lax NZ, Hepplewhite PD, et al. Relationship between mitochondria and alpha-synuclein: a study of single substantia nigra neurons. Arch Neurol. 2012;69:385–393. doi: 10.1001/archneurol.2011.2675. [DOI] [PubMed] [Google Scholar]
- 82.Mortiboys H, Aasly J, Bandmann O. Ursocholanic acid rescues mitochondrial function in common forms of familial Parkinson’s disease. Brain. 2013;136:3038–3050. doi: 10.1093/brain/awt224. [DOI] [PubMed] [Google Scholar]
- 83.Rockswold SB, Rockswold GL, Zaun DA, Zhang X, Cerra CE, Bergman TA, et al. A prospective, randomized clinical trial to compare the effect of hyperbaric to normobaric hyperoxia on cerebral metabolism, intracranial pressure, and oxygen toxicity in severe traumatic brain injury. J Neurosurg. 2010;112:1080–1094. doi: 10.3171/2009.7.JNS09363. [DOI] [PubMed] [Google Scholar]
- 84.Steer JH, Mann TS, Lo SZ, Inglis JJ, Yap HS, Henry PJ, et al. Early induction of uncoupling protein-2 in pulmonary macrophages in hyperoxia-associated lung injury. Inhal Toxicol. 2013;25:544–552. doi: 10.3109/08958378.2013.810679. [DOI] [PubMed] [Google Scholar]
- 85.Verweij BH, Muizelaar JP, Vinas FC, Peterson PL, Xiong Y, Lee CP. Impaired cerebral mitochondrial function after traumatic brain injury in humans. J Neurosurg. 2000;93:815–820. doi: 10.3171/jns.2000.93.5.0815. [DOI] [PubMed] [Google Scholar]
- 86.Halestrap AP. What is the mitochondrial permeability transition pore? J Mol Cell Cardiol. 2009;46:821–831. doi: 10.1016/j.yjmcc.2009.02.021. [DOI] [PubMed] [Google Scholar]
- 87.Crouser E. A rare glimpse behind the mask of sepsis-induced organ failures provides hope for an eventual cure. Am J Respir Crit Care Med. 2013;187:460–462. doi: 10.1164/rccm.201301-0107ED. [DOI] [PubMed] [Google Scholar]
- 88.Suh SW, Gum ET, Hamby AM, Chan PH, Swanson RA. Hypoglycemic neuronal death is triggered by glucose reperfusion and activation of neuronal NADPH oxidase. J Clin Invest. 2007;117:910–918. doi: 10.1172/JCI30077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Fuxe K, Rivera A, Jacobsen KX, Höistad M, Leo G, Horvath TL, et al. Dynamics of volume transmission in the brain. Focus on catecholamine and opioid peptide communication and the role of uncoupling protein 2. J Neural Transm. 2005;112:65–76. doi: 10.1007/s00702-004-0158-3. [DOI] [PubMed] [Google Scholar]
- 90.Boucher J, Kröger H, Sik A. Realistic modelling of receptor activation in hippocampal excitatory synapses: analysis of multivesicular release, release location, temperature and synaptic cross-talk. Brain Struct Funct. 2010;215:49–65. doi: 10.1007/s00429-010-0273-x. [DOI] [PubMed] [Google Scholar]
- 91.Mizuno T, Miura-Suzuki T, Yamashita H, Mori N. Distinct regulation of brain mitochondrial carrier protein-1 and uncoupling protein-2 genes in the rat brain during cold exposure and aging. Biochem Biophys Res Commun. 2000;278:691–697. doi: 10.1006/bbrc.2000.3859. [DOI] [PubMed] [Google Scholar]
- 92.Magleby KL, Stevens CF. A quantitative description of end-plate currents. J Physiol. 1972;223:173–197. doi: 10.1113/jphysiol.1972.sp009840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hestrin S, Nicoll RA, Perkel DJ, Sah P. Analysis of excitatory synaptic action in pyramidal cells using whole-cell recording from rat hippocampal slices. J Physiol. 1990;422:203–225. doi: 10.1113/jphysiol.1990.sp017980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Laughlin SB, de Ruyter van Steveninck RR, Anderson JC. The metabolic cost of neural information. Nat Neurosci. 1998;1:36–41. doi: 10.1038/236. [DOI] [PubMed] [Google Scholar]
- 95.Zhao QJ, Zhang XG, Wang LX. Mild hypothermia therapy reduces blood glucose and lactate and improves neurologic outcomes in patients with severe traumatic brain injury. J Crit Care. 2011;26:311–315. doi: 10.1016/j.jcrc.2010.08.014. [DOI] [PubMed] [Google Scholar]
- 96.Bayir H, Adelson PD, Wisniewski SR, Shore P, Lai Y, Brown D, et al. Therapeutic hypothermia preserves antioxidant defenses after severe traumatic brain injury in infants and children. Crit Care Med. 2009;37:689–695. doi: 10.1097/CCM.0b013e318194abf2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Nielsen N, Wetterslev J, Cronberg T, Erlinge D, Gasche Y, Hassager C, et al. Targeted temperature management at 33°C versus 36°C after cardiac arrest. New Engl J Med. 2013;369:2197–2206. doi: 10.1056/NEJMoa1310519. [DOI] [PubMed] [Google Scholar]
- 98.Salim A, Teixeira P, Ley EJ, DuBose J, Inaba K, Margulies DR. Serum ethanol levels: predictor of survival after severe traumatic brain injury. J Trauma. 2009;67:697–703. doi: 10.1097/TA.0b013e3181b5dcf2. [DOI] [PubMed] [Google Scholar]
- 99.Johnson VE, Stewart W, Smith DH. Axonal pathology in traumatic brain injury. Exp Neurol. 2013;246:35–43. doi: 10.1016/j.expneurol.2012.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Van Vleet TM, Chen A, Vernon A, Novakovic-Agopian T, D’Esposito MT. Tonic and phasic alertness training: a novel treatment for executive control dysfunction following mild traumatic brain injury. Neurocase. 2015;21:489–498. doi: 10.1080/13554794.2014.928329. [DOI] [PubMed] [Google Scholar]
- 101.Kim F, Nichol G, Maynard C, Hallstrom A, Kudenchuk PJ, Rea T, et al. Effect of prehospital induction of mild hypothermia on survival and neurological status among adults with cardiac arrest: a randomized clinical trial. JAMA. 2014;311:45–52. doi: 10.1001/jama.2013.282173. [DOI] [PubMed] [Google Scholar]
- 102.Rittenberger JC, Callaway CW. Temperature management and modern post-cardiac arrest care. New Engl J Med. 2013;369:2262–2263. doi: 10.1056/NEJMe1312700. [DOI] [PubMed] [Google Scholar]
- 103.Rostami E. Glucose and the injured brain-monitored in the neurointensive care unit. Front Neurol. 2014;5:91. doi: 10.3389/fneur.2014.00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Pandya JD, Pauly JR, Nukala VN, Sebastian AH, Day KM, Korde AS, et al. Post-injury administration of mitochondrial uncouplers increases tissue sparing and improves behavioral outcome following traumatic brain injury in rodents. J Neurotrauma. 2007;24:798–811. doi: 10.1089/neu.2006.3673. [DOI] [PubMed] [Google Scholar]
- 105.Dietrich WD, Bramlett HM. The evidence for hypothermia as a neuroprotectant in traumatic brain injury. Neurotherapeutics. 2010;7:43–50. doi: 10.1016/j.nurt.2009.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Sahuquillo J, Vilalta A. Cooling the injured brain: how does moderate hypothermia influence the pathophysiology of traumatic brain injury. Curr Pharm Des. 2007;13:2310–2322. doi: 10.2174/138161207781368756. [DOI] [PubMed] [Google Scholar]
- 107.Irimia A, Goh SY, Torgerson CM, Vespa PM, Van Horn JD. Structural and connectomic neuroimaging for the personalized study of longitudinal alterations in cortical shape, thickness and connectivity after traumatic brain injury. J Neurosurg Sci. 2014;58:129–144. [PMC free article] [PubMed] [Google Scholar]
