Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Nov 1.
Published in final edited form as: Phys Med Rehabil Clin N Am. 2011 Sep 25;22(4):577–602. doi: 10.1016/j.pmr.2011.08.002

Physical Medicine and Rehabilitation Clinics of North America 2011 The Pathophysiology of Concussions in Youth

Daniel W Shrey a, Grace S Griesbach b, Christopher C Giza c,d
PMCID: PMC3211100  NIHMSID: NIHMS327888  PMID: 22050937

SYNOPSIS

Mild traumatic brain injury, especially sport-related concussion, is particularly common among young persons. Consequences of transient pathophysiological dysfunction must be carefully considered in the context of a developing or immature brain, as must the potential for an accumulation of damage with repeated exposure over time. This review will summarize the underlying neurometabolic cascade of concussion with emphasis on the young brain in terms of acute pathophysiology, vulnerability, alterations in plasticity and activation, axonal injury, and cumulative risk from chronic, repetitive damage. The implications of these physiological changes will be discussed in the context of clinical care for the concussed youth, and areas for future investigation will be highlighted.

Keywords: traumatic brain injury, metabolic cascade, activation, plasticity, pediatrics

INTRODUCTION

Mild traumatic brain injury (TBI) or concussion is estimated to occur in millions of persons annually in the U.S. alone. The peak ages for these injuries are in adolescence and young adulthood, and sport-related concussions are particularly common among young persons [1]. While generally these injuries are self-limited, the consequences of even transient pathophysiological dysfunction must be carefully considered in the context of a developing or immature brain, as must the potential for an accumulation of damage with repeated exposure over a long period of time. This paper will review the underlying neurometabolic cascade of concussion with emphasis on the young brain in terms of acute pathophysiology, vulnerability, alterations in plasticity, axonal injury and risk for chronic cumulative deficits.

ACUTE METABOLIC CASCADE

Glutamate release and ionic disequilibrium

The post-concussive metabolic cascade has been well studied and characterized, both in animal models and in humans. As a result of mechanical trauma, neuronal cell membranes and axons undergo disruptive stretching, leading to temporary ionic disequilibrium [2]. As a result, levels of extracellular potassium increase drastically, andindiscriminate glutamate release occurs [3]. Glutamate release activates N-methyl-D-aspartate receptors, which leads to accumulation of intracellular calcium [46], causing mitochondrial respiration dysfunction, protease activation, and often initiating apoptosis [7, 8]. Elevated glutamate levels were also found to be significantly correlated with derangements in lactate, potassium, brain tissue pH, and brain tissue CO2 levels in human studies [9]. Additionally, sodium channel upregulation, fueled by ATPase proteins depending on glucose for energy, is observed following axonal stretch injuries [10].

Energy crisis and mitochondrial dysfunction

In combination, the cellular response to the above-mentioned ionic shifts and the downstream effects of the neurotransmitter release lead to an acute energy crisis. This occurs when, to restore ionic equilibrium, adenosine-triphosphate (ATP) -dependent sodium-potassium ion transporter pump activity increases, which augments local cerebral glucose demand [11]. Further metabolic demand is incurred by ATP-dependent sodium channel upregulation. This occurs in the face of mitochondrial dysfunction, leading cells to primarily utilize glycolytic pathways instead of aerobic metabolism for energy, and causing extracellular lactate accumulation as a byproduct [12]. This acidosis, caused by hyperglycolysis, has been shown to worsen membrane permeability, ionic disequilibrium, and cerebral edema [13].

Some evidence shows that the lactate produced by this process may eventually be utilized as a source of energy by the neurons once mitochondrial oxidative respiration normalizes; in fact, one study showed that in moderate to severe TBI the incidence of abnormally high levels of lactate uptake were seen in 28% of subjects [14]. The same study showed that patients exhibiting a higher rate of brain lactate uptake relative to arterial lactate levels tended to have more favorable outcomes compared to others with lower relative lactate uptake.

Alterations in cerebral blood flow

Cerebral blood flow changes following severe TBI have also been well-studied. A triphasic response to severe TBI has been characterized, specifically showing hypoperfusion on postinjury day 0, hyperperfusion on postinjury days 1–3, and vasospasm on postinjury days 4–15 [15]. Regarding mild TBI, some studies have shown that cerebral blood flow decreases immediately following the insult, and the amount of time it remains lowered seems to depend on the severity of the injury [16, 17]. Other studies, however, show no significant differences in CBF following mild TBI in subjects over 30 years of age [18]. In pediatric studies, CBF has been seen to increase during the first day following mild TBI, followed by decreased CBF for many days after [19, 20]. Data comparing cerebral blood flow in pediatric TBI patients has shown impaired autoregulation in 42% of moderate and severe and 17% of mild injuries [21]. In severe TBI, impaired autoregulation was associated with worsened long-term outcomes [22].

VULNERABILITY TO SECOND INJURY AND SECOND IMPACT SYNDROME

Post-concussive physiological changes have been shown to increase the brain’s vulnerability to further injury, particularly in cases where a second concussive injury is sustained within days of the first. This phenomenon can lead to more severe and permanent deficits. Numerous studies, both in animal models and in humans, support the concept of post-concussive vulnerability, prompting the development of many sets of return-to-play guidelines [2328]. Research has shown that having a history of concussion increases an individual’s probability of having a future concussion as well as prolonging the duration of significantly abnormal cognitive functioning [29]. These data support increased awareness of post-concussive vulnerability and suggest adherence to return-to-play guidelines may be helpful.

Additionally, an extreme entity termed “second impact syndrome” has been postulated, where an individual experiencing a second mild TBI within days of the first can experience diffuse cerebral swelling and catastrophic deterioration [30, 31]. Second impact syndrome remains somewhat controversial in definition and explanation [27, 3133], with most of the literature addressing its existence being case reports. The pathophysiology of second impact syndrome is thought to result from the combined effects of increased intracranial blood volume (secondary to loss of cerebral blood flow autoregulation) and post-traumatic catecholamine release, leading to catastrophic cerebral edema, generally in the absence of major hematomas or other space-occupying lesions [3335]. McCrory et al. proposed diagnostic criteria in 1998 to facilitate more consistent analysis of potential cases of second impact syndrome, as well as to diminish the bias involved in case-reporting [32]. After applying these criteria, many of the previously reported cases of second impact syndrome were found to fall short of diagnosis. Nonetheless, there is recognition of a syndrome of malignant cerebral edema after mild TBI, and multiple reviews and case series of second impact syndrome have been published, [33, 36, 37]. Interestingly, a similar syndrome has been described in individuals harboring a calcium channel mutation (CACNA1A) associated with familial hemiplegic migraine [38].

Experimental

Numerous basic science studies have investigated the consequences of sustaining two closely timed brain injuries as compared to a single insult, characterizing the effects on brain damage and cognitive deficits [2326, 39]. As previously described, neurons undergo significant changes in metabolism, blood flow, and energy usage following a single TBI. These changes in cellular function contribute to transient dysfunction after injury, but in mild TBI, cellular recovery eventually occurs, typically with minimal anatomical changes/damage [40]. Metabolic derangements in NAA, ATP, and ATP-to-ADP ratios after concussive brain injuries in rats have been studied. Results showed that 2 concussive injuries separated by 3 days showed derangements significantly worse than either a single injury or 2 injuries with 5 days separation [41]. In a more recent study, Vagnozzi et al. measured other biochemical markers of mitochondrial dysfunction in rats sustaining two mild TBIs, showed comparable findings. Another notable result was that a second impact at 5 days after the first showed metabolic changes equivalent to the first impact but not more severe [25]. Sustained oxidative and nitrosative stresses have also been shown to follow a similar pattern with peak derangements noted with a second impact occurring 3 days after the first [24].

These results corroborated those of an earlier study from 2005 by Longhi et al., who compared mice with one concussive event or two concussive events one, three, five, or seven days apart with control mice that underwent sham injuries. This study showed significant cognitive deficits in mice concussed twice at intervals of 3 and 5 days but not in those concussed only once, or concussed twice but at 1 or 7 day intervals. This supports the notion of a defined interval following an initial insult during which there is increased propensity for greater deficits should a second trauma occur. This study also showed a significantly increased degree of traumatic axonal injury mice sustaining a second injury 3 days after the first injury when compared to mice with only a single concussion [39].

Other studies have focused on histological and cognitive changes after mild TBI [26, 42]. It has been shown that experimental closed head injury in juvenile rats can cause cognitive deficits in the absence of gross pathology or high mortality rates. However, a second injury performed one day after the first caused increased memory impairment as well as neuropathological changes of increased axonal injury and astrocytic reactivity [26]. This study is also the only experimental study of repeated mild TBI conducted in immature animals, thus targeting the age group at highest risk for such injuries in humans. Another study showed impairment in Morris water maze following three sequential concussive head traumas causing transient loss of consciousness, but these injuries also caused contra-coup neuropathological changes compared with non-concussed controls [42].

Human studies

Magnetic resonance spectroscopy has been used to assess cellular changes after sustaining TBI [23, 43, 44]. Vagnozzi, et al. [23], using voxels in the white matter of the bilateral frontal lobes immediately adjacent to the cortical-subcortical junction, assessed the ratio of N-acetylaspartate (NAA) to creatine (Cr) specifically in athletes after concussion. Concussed athletes showed a decrease (−18.5%) in the NAA-Cr ratio on post-injury day 3 compared to age-matched, non-concussed controls (1.80 ± 0.04 vs. 2.15 ± 0.1; P<0.001), with return to normal values by 30 days post-concussion. A second concussive event prolonged the time of NAA:Cr normalization by 15 days, with NAA:Cr values remaining significantly below normal (1.82 ± 0.1) at 30 days, and return to normalcy noted only at 45 days (2.07 ± 0.1) [23].

Data described in the above studies, coupled with the lack of consensus regarding management of concussion based on clinical evaluation alone, would support supplementing clinical assessment by using biomarkers. This would add more objectivity and allow for better defined return-to-play recommendations. The necessity of including such data in gauging severity of TBI and recovery stems from the demonstration that an absence of clinical signs and/or symptoms does not always coincide with completed brain metabolic recovery. Conversely, it is still unclear whether metabolic alterations in the absence of measurable clinical impairment are indicative of an underlying vulnerable state. A potentially valuable approach might be to identify clinical measures that closely reflect the underlying metabolic state in a subset of athletes, as it is currently impractical to propose routine use of MRS for all concussions. Ideally, the approach to management of a concussed individual would then involve monitoring of clinical status in all and metabolic biomarkers in a selected subset of athletes.

IMPAIRED/ABNORMAL ACTIVATION AFTER TBI

Experimental

Experimental brain injury models have indicated that brain activation is altered for several weeks after TBI. Changes in excitability and circuit function have not only been observed in more severe brain injury but also following mild TBI [45, 46]. Anatomical changes after trauma along with subtle changes in neuronal properties will influence neuronal excitability. These post-injury changes in brain activation have an array of consequences ranging from alterations in synaptic plasticity [46], axonal sprouting [47] and excitatory-inhibitory imbalance [48]. These underlying physiological changes may then manifest phenotypically as cognitive impairments and/or increased risk of seizures/epilepsy [49].

Following a brain injury, metabolic alterations occur that result in subacute hypometabolism. The duration of this metabolic disturbance is thought to be associated with the severity of the injury [50]. These metabolic changes are likely to influence brain activation [5154]. Changes in excitability also have an effect on calcium regulation and thus on molecular markers of plasticity such as neurotrophins. Animal research indicates that neurotrophin expression, such as brain-derived neurotrophic factor (BDNF), is regulated by neural activity [55, 56]. In turn, alterations in BDNF will also influence brain activation given its role on synaptic facilitation [57] and neurotransmitter release enhancement [5860]. For example, BDNF levels normally increase in response to exercise [61]; however, this is not observed if exercise occurs during the first days after a fluid-percussion injury (FPI) [62]. Among these lines, cortical stimulation to the brain following FPI has been observed to elicit a metabolic response that may act as a secondary injury by increasing cortical degeneration [63]. Metabolic and ionic disturbances will also affect function of the N methyl D-aspartate receptor (NMDAR), which is a key molecular player in neuronal activation and allows for long-term potentiation (LTP), a cellular correlate of learning. As a glutamate receptor, the NMDAR plays a major role in neuronal excitability. It should be noted that the response to injury, such as the period of metabolic depression, differs between adult and developing animals [5, 64, 65].

Changes in brain activation can also have an effect on the regulation of the hypothalamic pituitary adrenal (HPA) axis. Efficient HPA regulation is necessary for neuronal vitality and function, thus its dysregulation can have profound effects on synaptic plasticity as well as cognitive and affective well being. Glucocorticoid release is regulated by the hypothalamus, a region receiving multiple suprahypothalamic inputs with notable projections from the limbic system. Alterations in function and connectivity are likely to exert a substantial influence on HPA regulation. Glucocorticoid receptors are widespread throughout the brain and are predominantly expressed within the hippocampus [66, 67]. Excitatory changes within the hippocampus will thus have an effect on glucorticoid negative feedback. Recent findings from adult rodent studies indicate that there is a hyper-responsiveness to stress following a mild FPI. This hyper-responsiveness was observed during the first postinjury weeks [68]. These increases are indicative of hypothalamic disruptions in neuronal circuitry. A heightening of the stress response may be particularly concerning in the developing brain. Neonatal or pubertal stress has a pronounced effect on adult behavior and brain plasticity [6971]. Along these lines, corticotropin releasing hormone mRNA has been shown to increase after TBI [72, 73]. In addition to the heightened stress response, animal studies, utilizing different injury models, have indicated other TBI-induced changes in HPA regulation [74, 75].

Human studies

As shown in animal models of injury, neurochemical and metabolic disruptions are also observed in humans. Metabolic disruptions after experimental TBI extend over a period of days and normalize after approximately 10 days [11, 12], while changes after moderate-severe human TBI may last weeks or even months [76]. In addition, the consequences stemming from a TBI include white matter damage and changes in activation patterns. These alterations are likely to contribute to the long-term cognitive impairments that are observed after TBI. These range from acute memory problems to difficulty with higher executive functions such as strategy switching and planning. A functional MRI study of individuals with mild or moderate brain injury found neural activity alterations a year after injury. In particular, disruptions in catecholaminergic circuitry, which is associated with working memory functions, were observed [77]. Alterations in activation after mild brain injury are also associated with axonal damage, but this is generally not apparent with computed tomography [78]. Indeed subtle white matter abnormalities are observed in patients with persistent postconscussive symptoms after suffering from a mild TBI using Diffusion Tensor Imaging (DTI; see below) [79].

As indicated above, changes in activation patterns are likely to have an effect on glucocorticoid regulation. Acute activation of the HPA axis occurs initially after brain injury as a protective response that modulates the immune/inflammatory response and increases metabolic substrate availability [80]. Clinical studies have demonstrated elevations in cortisol acutely after brain trauma and these elevations are correlated with the duration of coma and recovery after head trauma [81]. As for the subacute period, studies suggest that plasma cortisol levels are dependent on the injury severity. Cortisol tends to increase after mild to moderate brain injury in the early post-injury period, whereas it is depressed after a severe injury [82, 83]. Although structural pituitary damage may be dependent on the severity of the injury [84], pituitary abnormalities have been reported in mild to moderate injuries. MRI studies indicate that 37% of mild TBI adult patients, with a Glasgow Coma Score ranging from 13–15, had structural pituitary abnormalities [85]. A higher rate of pituitary abnormalities was found in a pediatric study looking at functional measures. Basal cortisol levels were significantly reduced in this population [86]. It still remains to be answered if there is an age dependent vulnerability of the HPA axis.

ALTERED PLASTICITY AND DEVELOPMENT

Experimental

Brain injury leads to alterations in molecular substrates of synaptic plasticity. Of particular interest are the effects on proteins such as BDNF and the NMDAR, which are strongly linked with synaptic strengthening and play a significant role in experience-dependent plasticity [87, 88]. Because the young brain is undergoing developmental changes, injury-induced alterations of molecular markers of plasticity may not only affect injury outcome, as it has been shown in adults, but also deviate developmental processes from their normal trajectory.

It is during the developmental period that the brain is more susceptible to environmental influences. This has been illustrated in enriched environment (EE) studies. These studies indicate that structural changes are more likely to occur in a developing rat as a response to environmental stimulation when compared to an adult rat [8991]. However, when a developing rat is exposed to an EE after a FPI, the beneficial effects indicative of neuroplasticity, such as cortical thickening, expanded dendritic arborization and cognitive enhancement, are not observed [92, 93]. The cognitive benefit that is usually found after EE exposure is only found if the period of enrichment is delayed after the injury [94]. Interestingly, this failure to enrich after developmental TBI is observed in spite of no significant cell death [95]. This demonstrates that mechanisms driving neuroplasticity are affected after TBI and may lead to the failure to appropriately respond to environmental stimulation. In accordance with this idea, changes in NMDAR composition have been observed after a FPI in young rats [96]. In addition, alterations in molecules that facilitate synaptic plasticity, such as BDNF, are likely contributors to the post-TBI failure of EE.

Adult TBI studies have shown that expression of brain-derived neurotrophic factor (BDNF) is acutely altered in different models of TBI [97100]. BDNF mRNA is dynamically regulated during postnatal development in various CNS structures and its levels are high and unstable compared to the mature brain [101103]. Therefore, it is plausible that the effects of TBI on BDNF are age dependent. Indeed, levels of BDNF were observed to be elevated after a FPI in developing rats when compared to adult FPI [104, 105]. These increases in BDNF were most pronounced in areas remote from the injury site and may be indicative of synaptic remodeling. Reorganization after brain injury has been proposed as a mechanism to facilitate recovery [106108] and has been observed in animals after brain injury [59, 93, 109112].

A substantial increase in neuronal connectivity occurs during the postnatal period, as a response to functional and environmental demands. During this period, the organization of fiber projections takes place through regionally selective outgrowth or selective elimination of projections [113, 114]. These ongoing maturational processes emphasize the significance of TBI studies in this age range. These studies suggest that following TBI, future developmental plasticity may be compromised, in that neuroplasticity shifts from responding to experience-dependent adaptation to that of restoration/compensation to a pre-injury status.

Human studies

Data obtained from human TBI studies indicates that the capacity of the brain to compensate for injury is largely dependent on the state of cerebral maturation. For example, children younger than four years have a worse outcome compared to older children [115, 116]. Paradoxically, although children may be more behaviorally resilient to injury, cognitive deficits may not be noticeable until the child is exposed to a more demanding environment. Thus, deficits in attention, memory and verbal abilities may only become apparent later in development; these impairments after pediatric TBI may also manifest as an increased likelihood of needing special education programs [117119].

The brain is rapidly developing during the first years. Events refining neural connectivity are actively taking place through processes such as myelination, dendritic changes and synaptogenesis [120]. As suggested by the animal literature, TBI in young children jeopardizes ongoing developmental processes [121, 122]. Studies by Anderson indicate that executive functions, such as information processing and the ability to suppress impulsive behaviors, are particularly affected in those children who suffered a TBI before the age of 3 [116, 123]. Interestingly deficits in executive functions were observed regardless of injury location [124]; thus suggesting that the alteration of neural networks due to injury had an effect on skills that are acquired at a future developmental period. With the exception of attentional control, it was the skills that were in the process of being acquired at the time of injury that were more likely to show impairments. A recent study of young adults that had suffered from a TBI during childhood indicates that these impairments may be long lasting, particularly following a severe TBI. In these individuals quality of life and employment opportunities were reduced [125].

TIMING OF RETURN TO ACTIVITY

As discussed above, there is substantial evidence that neural activation and plasticity are altered after developmental TBI. It is also known that physiological neural activation can promote recovery, while excessive activation may exacerbate cellular damage. These neurobiological principles, then, underlie the clinically relevant determination of the optimal timing for return to activity following TBI/concussion.

Experimental

Ideally exercise should facilitate the capacity of the brain to compensate for insults, thus resulting in a better outcome. Numerous studies have shown that exercise, particularly of a voluntary nature, increases markers of neuroplasticity and promotes neurogenesis [61, 126131]. However, this may not always be the case if particular considerations are not taken into account when implementing exercise in the post-concussive setting.

Animal studies have shown that the beneficial effects of exercise are only found if exercise is delayed after TBI. In these studies, rodents underwent a mild FPI and were allowed to exercise for a week, starting on the day of the injury. Not only did these rats fail to show an increase in BDNF and other target proteins, but also presented cognitive deterioration when compared to their sedentary counterparts [62]. Moreover, experimental findings suggested that premature exercise disrupts restorative processes. Proteins associated with plasticity that were increased as a result of TBI, in areas remote from the site of injury, were reduced when rats were acutely exercised [104]. Compensatory and/or restorative responses may be observed in areas of the brain that have endured a lesser amount of harm [106108, 132135].

In contrast to acute postinjury exercise, when exercise was delayed after an FPI, increases in BDNF were observed. This exercise-induced increase in BDNF was associated with enhanced hippocampal dependent learning and memory [136]. A cognitive benefit is typically observed as a result of voluntary exercise [137]. In addition to BDNF, exercise increases other proteins that enhance neural function and cognitive performance. Some of these proteins have been found to increase with exercise after experimental brain injury. For example calcium-calmodulin-dependent protein kinase II (CaMKII) and cyclic-AMP response-element-binding protein (CREB) are elevated when delayed postinjury exercise. Both of these have been shown to facilitate long-term potentiation (LTP), a key cellular mechanism underlying synaptic plasticity and learning. In addition animal studies suggest that this time window is dependent on the particular characteristics of the injury; for example, the necessary delay for exercise to be effective after a FPI is severity dependent [138].

Although it is still unknown why the injured brain does not respond to acute postinjury exercise it is plausible that untimely exercise may divert cerebral metabolism from needed functions such as energy restoration and production of synaptic plasticity molecules, by introducing an increase in metabolic demand at a time when the brain is energetically compromised. TBI results in mitochondrial dysfunction and decreases in blood flow that compromise neuronal functioning and signaling. These disruptions in cellular function along with injury-induced changes in activation are likely to interfere with the effects of exercise.

In addition, as indicated above, TBI disrupts the regulation of stress hormones, and may heighten the stress response for the first post injury weeks. Glucocorticoids, which are known to suppress levels of BDNF and other key proteins [107, 139143], may be elevated after injury and contribute to the undesired effects of early post-injury exercise. In addition, a hyper-response to stress is likely to magnify exercise’s regular metabolic effects. Exercise by itself places an energetic demand and moderately increases levels of corticosteroids [144, 145]. Along these lines it should be noted, that some forms of exercise elicit a stronger stress response. Different forms of exercise will involve distinct motivational and fatigue characteristics. Thus certain exercise regimens with stronger stress responses may be particularly counterproductive during the early post-injury time period.

Human studies

As in animal studies, neuroprotective qualities have been associated with exercise in humans. There is a wealth of studies indicating the beneficial effects of exercise on cognitive function and mental health [146150]. In children, exercise is associated with improved academic performance and executive functioning [151154]. Exercise has also been found to decrease headaches [155, 156], which are one of the main symptoms reported after mild TBI in both children and adults.

The effects of exercise have also been explored in the TBI population. A retrospective study that relied on self-reports found that postinjury exercise decreased negative mood states in adults [157]. Affective disorders are prevalent in the TBI population [158, 159]. Another study found that the practice of aquatic activities, at least 12 months post TBI, decreased the incidence of negative moods [Driver 2004]. Although most of the exercise studies assessing postconcussive effects have been in adults, it is also likely that the benefits of exercise will be present in a younger population. A review of different trials with post TBI subjects ranging in age from 3 to 20 years found a short-term improvement in self-esteem [160]. Other studies have found that exercise lessens post-concussive symptomatology, such as headaches, dizziness and fatigue [161]. This study utilized gradual and controlled exercise rehabilitation in a pediatric TBI population that presented symptoms 4 to 18 weeks post TBI.

Exercise studies in the TBI population have included a wide range of post-injury periods, but few offered observations acutely after injury. Among these, a retrospective cohort study by Majerske found that high levels of cognitive and physical activity during the early postinjury period had a negative effect on cognitive function and were associated with worsened concussive symptoms and performance on cognitive testing [162] [Majerske J Athl Training 2008]. The occurrence of negative mood states and cognitive impairments may be associated with neuroendocrine alterations that are found in the early post-TBI weeks, such as elevated plasma cortisol levels [83].

These findings indicate that premature neural activation alone, without a repeated injury, can have a deleterious effect. Thus it is not only the risk of a repeated concussion that is of concern when addressing the return-to-play, but also the potential deleterious effect of excessive, early neural activity. It should be noted that when a second concussion occurs the brain is more susceptible to damage due to ongoing pathology from the first injury [39, 163, 164]. Because of these concerns, consensus guidelines for the reinstatement of physical activities following TBI [165167], recommend avoiding activities such as physical and cognitive exertion until postconcussive symptomatology has resolved. Determining the optimal time for return-to-activity can be very challenging in the sports or military setting. Having fixed waiting guidelines may be too conservative or not sufficient, because injury characteristics and recovery vary greatly between patients. However symptom resolution may not always be self-reported accurately, nor may it reflect full cerebral or neurocognitive recovery [23, 168, 169]. The timeline for effective exercise therapy as well as return to physical activities is dependent on the characteristics of the injury as well as previous injury history and thus should be tailored on an individual basis. Due to these concerns it is important to understand the physiological and neurobehavioral correlates that underlie the injury. Unfortunately the literature regarding therapeutic exercise is largely based on anecdotal data and randomized studies on TBI patients are sparse, indicating a pressing need for further rigorous investigations.

AXONAL INJURY

Experimental

It is well known that biomechanical forces applied to neural tissue results in dysfunction and damage to axons [170172]. Changes in axonal integrity and function have been described in experimental models of mild TBI [39], including a recently described model of repeat concussive injury in the juvenile rat [173]. In this juvenile model, the degree of axonal damage and glial reactivity were amplified when two closed head injuries were experienced 1 day apart. Behaviorally, a single mild TBI caused a decrement in working memory that was worsened by the combined injury.

Another consideration when investigating axonal damage during development is the fact that myelination is ongoing in many brain regions throughout cerebral maturation. There is evidence that unmyelinated fibers may be more vulnerable to TBI, as demonstrated by Reeves, et al [174]. In that study, midline FPI resulted in a loss of compound action potentials through the corpus callosum, and, while recovery of the myelinated fibers occurred over time, persistent deficits in the unmyelinated peaks were seen. This data suggests that unmyelinated axons, and thus immature axons, could be more vulnerable to biomechanical injuries.

Lastly, imaging of animal TBI models provides insight into axonal injury. For example, Diffusion Tensor Imaging (DTI), an advanced MRI technique that measures directional diffusion as a marker of white matter fiber integrity, has been studied following experimental controlled cortical impact injury. Changes in DTI signal have been correlated with histopathological findings [175, 176].

Human studies

In the adult brain, there is substantial pathological evidence for axonal injury after severe TBI [177, 178], but understandably only a few such reports of axonal damage after milder, concussive injury, which were in older persons [179, 180]. Advances in noninvasive neuroimaging, particularly DTI, have provided new insight into white matter damage or dysfunction occurring after TBI, including in the pediatric and young adult population [181184]. Fractional anisotropy (FA) is a measure of the directionality of water diffusion, with a value of 1 being the highest, and a value of 0 indicating diffusion in equally in all directions (essentially no directionality).

In children with moderate-severe TBI compared to orthopedic controls, DTI showed abnormalities in frontal white matter 3 months post-injury. These abnormalities, such as decreased FA, were predictive of long-term global outcomes [182]. Similar reductions of FA were reported in the corpus callosa (CC) of adolescents after moderate-severe TBI, which was also correlated with reduced CC NAA on MRS [185].

Studies of DTI after mild TBI/sports concussions have been limited to older adolescents and adults (for review of DTI methods in general, see 186]. An early pediatric DTI study compared control subjects to those with mild and moderate TBI (average age 15.1±2.3 years, post-injury time 8.2±2.2 months). This study showed lower FA values in moderate TBI children and neurocognitive impairments in the combined mild-moderate TBI group [181]. Specifically in the supracallosal region, both mild and moderate groups separately showed significantly reduced FA compared to controls. Wilde, et al. [183] measured DTI in adolescents within the first week after concussion, and interestingly showed increased FA in corpus callosum, which correlated strongly with the clinical scores on the Rivermead Post-Concussion Symptom Questionnaire (RPCSQ). This increased FA was postulated to be indicative of early axonal swelling, which would favor diffusion along the direction of the callosal fibers. Using multimodal imaging combining DTI and magnetoencephalography (MEG), a group of young military and civilian adults with post-concussion symptoms and normal conventional neuroimaging were studied. This study showed areas of decreased FA in subcortical white matter underlying areas of focal slowing on MEG [184] and supported the utility of this combined imaging methodology to distinguish between mild TBI patients and controls.

It is likely that both DTI and MRS of white matter fibers will yield greater insight into post-concussion axonal pathobiology. Such noninvasive scans may be readily conducted in humans, and translational work investigating MR imaging and histopathology in experimental animal models will greatly aid in the elucidating mechanisms underlying abnormal white matter signals on advanced MRI sequences. Further critical work is also required in establishing clinical correlates of these white matter abnormalities, determining the longitudinal time course of axonal injury and distinguishing differences in traumatic axonal injury between the pediatric and adult brain.

CHRONIC TRAUMATIC ENCEPHALOPATHY AND LATE RISK OF DEMENTIA

Chronic traumatic encephalopathy is a progressive neurodegenerative disease found in some individuals subjected to repetitive mild TBI. Neuropathologically, it can be described as a tauopathy of the brain manifesting as neurofibrillary tangles throughout most of the brain with a relative paucity of beta-amyloid deposition [187].

Experimental

Single and recurrent mild TBI and the effect on amyloid proteins associated with dementia have been extensively studied in animal models. The majority of research has focused on Aβ amyloid accumulation and its correlation with cognitive impairment. Increased levels of amyloid precursor protein (APP) have been found in animals undergoing recurrent TBI [188], as well as increased levels of β-secretase [189] and γ-secretase enzymes [190], which are both known to convert APP into neurotoxic Aβ peptides. Likely related to these findings, Aβ1–40 and 1–42 levels have also been found to be significantly increased in the hippocampal neurons of PDAPP transgenic mice overexpressing mutant human APP who are subjected to cortical impact brain injury compared to non-injured transgenic mice. The injured hippocampi showed substantial exacerbation of neuronal death, postulating that there is a mechanistic link between brain trauma, Aβ levels, and neuronal cell death [191]. With repetitive mild TBI in Tg APP695swe (Tg2576) transgenic mice that overexpress mutant APP, accelerated deposition of Aβ plaques and earlier onset of cognitive impairment are also seen [192]. Research with non-transgenic mice has shown that after controlled cortical impact, accumulation of mouse Aβx-40 peptide was noted after 1 day, with increasing levels of almost 120% seen by 3 days, and normalization of levels by 7 days [193]. Other evidence supporting the involvement of Aβ plaque formation in long-term outcomes comes from research done by exposing pigs to inertial brain injury, which was found to cause long-term (up to 6 months post-injury) accumulation of APP, beta-site APP cleaving enzyme (BACE), pre-senilin 1, and activated caspase in the axons of injured neurons. The accumulation of these factors is believed to be due to impaired axonal transport due to trauma, and it may lead to APP proteolysis and Aβ formation within the axonal membrane compartment [194]. Post-concussive Aβ plaque formation may help to explain the increased risk of Alzheimer disease after mild TBI through acceleration of the postulated amyloid cascade [195].

Gross pathology

Individuals with a history of multiple episodes of mild TBI may have one or a number of stigmata associated with repetitive concussion. Often seen are such findings as an anterior septum pellucidum cavum, as well as posterior fenestrations in the ventricular system [196]. Individuals with septum pellucidum cavum may also develop recurrent obstructive hydrocephalus due to abnormalities in CSF flow, requiring ventricular fenestration or ventriculo-peritoneal shunt placement [197]. Other gross pathology includes atrophy of the fronto-temporal cortex, medial temporal lobe, hypothalamic floor, and mammilary bodies, as well as enlargement of the third and lateral ventricles, hippocampal sclerosis, and pallor of the substantia nigra. A reduction in total brain volume may be seen, with atrophy noted in the cerebrum, diencephalon, basal ganglia, and brainstem [187].

Microscopic pathology

A number of different microscopic findings are seen with repetitive chronic mild TBI, including tau-positive neurofibrillary tangles, astrocytic/glial tangles, and neurites of spindle and threadlike shapes. Deposition of beta-amyloid occurs in less than half of cases, according to one retrospective study [187]. Additionally, recent attention has been drawn to the potential involvement of Transactive Response (TAR) DNA binding protein-43kDa (TDP-43) in the pathophysiology of chronic traumatic encephalopathy [198]. Diffuse cerebral white matter degeneration has long been known to occur with severe dementia following head injury [199]. Multifocal axonal injury with damage in the corpus callosum and the fornices has been rarely described after single mild TBI in patients who died shortly after injury due to non-neurological complications [179]. Gross fornix degeneration has also been noted in similar patients [200].

Tau-based protein deposits

Historically, neurofibrillary tangles, mostly comprised of tau protein, have been regularly seen in the brains of subjects with a history of multiple mild traumatic brain injuries. The exact mechanism that leads to their accumulation is not known. As previously mentioned, shearing forces acting on neuronal axons leads to increased cell membrane permeability, ionic disequilibrium, and often triggers intracellular apoptotic enzymes, which may initiate and later aggravate formation of neurofibrillary tangles via tau truncation, phosphorylation, and misfolding [171, 201, 202]. Also contributing to their production and accumulation is cytoskeletal element breakdown, notably microtubules and microfilaments [203].

Neurofibrillary tangles are seen predominantly in the superficial cortex (layer II and the upper portion of layer III), mostly in frontotemporal and insular distributions [204]. They are generally found in patches, notably in the deeper portions of the sulci as well as subpial, periventricular, and perivascular areas [205, 206]. They are often seen with neuritic threads, seen in the subcortical nuclei/basal ganglia, hippocampus, brainstem nuclei, and cerebral cortices [207], with notable cortical involvement of the frontal and temporal lobes, as well as the insula [187]. According to McKee et al., both neurofibrillary and glial tangles were found in the medial temporal lobe, diencephalon, brain stem, and basal ganglia, as well as subcortical white matter of subjects with chronic traumatic encephalopathy. The perivascular distribution of neurofibrillary tangles suggests that mechanical disruption of the cerebral microvasculature may play a role in their deposition [187]. These distributions remarkably contrast those of Alzheimer disease, which are generally seen in a more uniform cortical distribution, mostly in cortical layers III and V, without perivascular predominance.

Extension of the tau-based neurofibrillary tangles from the isolated perivascular spaces to broader, patchy distributions is not completely understood, however, some evidence suggests misfolded tau proteins are able to propagate out of one cell and into another, similar to prions. This phenomenon has been observed in vitro and is described as extracellular tau aggregate being taken up by cultured cells and internalized, leading to fibrillization of new tau proteins intracellularly via displacement of tubulin and co-localization with dextran [208].

Beta-amyloid (Aβ) deposits

Neuritic beta-amyloid plaques are seen in slightly less than half of all cases of chronic traumatic encephalopathy. Their distribution differs markedly from those found in Alzheimer disease, as subjects with multiple concussions demonstrate diffuse plaques with less density than the abundant plaques seen in Alzheimer disease [205]. Although the pathogenesis of beta-amyloid deposits is not entirely clear, amyloid precursor protein has been found to accumulate in the axonal bulbs of injured neurons following diffuse axonal injury, as seen in mild TBI, most commonly in subcortical and deep white matter [209]. Amyloid precursor protein is then likely cleaved to form beta-amyloid. There is conflicting evidence regarding the exact role played by beta-amyloid in pathogenesis vs. neuroprotection in injured neurons [209, 210].

TAR DNA binding protein-43kDa (TDP-43)

A recent study reported neuropathological evidence for the theoretical epidemiological linkage between head injury and amyotrophic lateral sclerosis. A small number of cases with chronic traumatic encephalopathy and severe motor neuron disease were found to have TDP-43 deposits in the brain as well as in the anterior horns of the spinal cord [198]. The conclusions drawn from these results, most notably a link between repetitive mild TBI and amyotrophic lateral sclerosis, remain controversial [211, 212].

CSF markers of repetitive head trauma

Short term biochemical aberrations in the face of head trauma have been investigated. CSF markers of repetitive head trauma (neurofilament light protein, total tau, glial fibrillary acidic protein, phosphorylated tau, and β-amyloidprotein 1–40 and 1–42) have been studied in amateur boxers at 7–10 days and 3 months post-fight and compared with healthy, non-athletic controls. Neurofilament light protein, total tau, and fibrillary acidic protein were found to be increased shortly after fights compared to 3 months later in boxers, with the increase dramatically higher among those receiving more punches or more high-impact hits to the head. Compared to controls, the boxers had significantly higher levels of neurofilament light protein and glial fibrillary acidic protein 7–10 days after a fight. Neurofilament light protein was the only CSF marker found to be significantly elevated 3 months after a fight in boxers compared to controls [213].

Other CSF biomarkers have been identified and found to be altered long-term in those with repetitive head trauma. Zetterberg et al. compared CSF levels of S-100B, brain-derived neurotrophic factor (BDNF), heart-type fatty acid binding protein (h-FABP), glial fibrillary acidic protein (GFAP), and neuron-specific enolase using biochip array in amateur boxers after 2 months of boxing abstention compared to healthy controls with no history of head trauma. They found that only neuron-specific enolase was significantly elevated, while the remainder of the CSF markers tested showed no differences between the boxers and controls [214]. Finding no significant differences in CSF tau concentrations between resting boxers and controls supports CSF tau as a biomarker of acute concussive injury and not chronic repetitive mild brain trauma. Alterations in neurofilament light protein were not mentioned in the 2009 study.

Genetic susceptibility to chronic traumatic encephalopathy

In addition to previously mentioned neuropathological similarities between chronic traumatic encephalopathy and Alzheimer disease, the genetics predisposing to both disorders also seem to have significant overlap. It is well known that apolipoprotein E is a susceptibility gene for late-onset familial and sporadic Alzheimer disease, and that possessing the ε4 genotype leads to dose-dependent hastening of the onset of Alzheimer disease [215217]. The development of chronic traumatic encephalopathy has also been linked to the ε4 genotype [218]. Having the same Apolipoprotein E ε4 genotype also predisposes the individual to incurring significantly more severe chronic traumatic encephalopathy compared to those without the ε4 genotype, given the same degree of chronic TBI [219]. It is unknown whether possessing the Apolipoprotein E ε2 genotype is protective in chronic traumatic encephalopathy, as it seems to be in Alzheimer disease [220]. The implications of these findings support the theory that chronic traumatic encephalopathy, as well as Alzheimer disease, share a complex combination of both environmental and genetic risk factors. Future consideration should be given to the possibility of genetic counseling for professional athletes likely to encounter repeated head trauma and their likelihood of developing an encephalopathic state.

CONCLUSIONS

Our understanding of the phenomenon of concussion has been shaped significantly by experimental work in animal models, as well as extrapolation of physiological measurements from humans with more severe TBI. This review covers three main post-concussive periods: 1) the acute neurometabolic cascade, 2) the subacute phase of altered neural activation and axonal disconnection, and 3) the chronic accumulation of insults that may lead to permanent impairments. The acute neurometabolic cascade involves indiscriminate neurotransmitter release, ionic disequilibrium, metabolic crisis (with energy depletion and oxidative stress) and cytoskeletal damage. These pathophysiological alterations also occur after mild TBI and create a state of vulnerability to repeated injury. In animals, these acute neurometabolic perturbations may take 7–10 days to recover fully. In humans, abnormal metabolic patterns seen on MRS after mild TBI may require up to 4 weeks to recover. Beyond the acute changes, concussive brain injury is followed by a period of impaired neuroplasticity and axonal disconnection. Both of these phenomena may have significant implications for recovery when they occur and/or recur in the immature brain, as they may result in a slowed developmental trajectory, with persistent or emergent neurocognitive deficits that only fully manifest over time. Following mild TBI, experimental evidence indicates that not only is the concussed brain less responsive to physiological neural activation, but it may also be more vulnerable to premature forced activation which can act as a secondary stressor to exacerbate dysfunction and neural damage. These findings correlate loosely with clinical data suggesting that repeated concussions or excessive levels of post-injury activity lead to longer duration and greater severity of post-concussive symptoms. Together, these preclinical and clinical data form the basis for consensus recommendations of delayed return-to-activity and return-to-play. Finally, there is growing evidence for chronic, cumulative neurobiological impairment, centered on abnormal protein deposition, delayed cell death and axotomy and resultant cognitive and behavioral decline.

Knowledge of these cellular mechanisms is important for understanding the distinctions of concussions as they occur in children and adolescents. First, the young brain may be biologically more vulnerable to repeat mild TBI, through impairments in neural activation/plasticity and increased sensitivity of developing axons to biomechanical forces. Second, the population of young children exposed to contact sports is relatively unselected genetically, and thus, genetic risk factors for worsened acute symptomatology have not yet been weaned out, as they may be in more elite collegiate or professional adult athletic populations. Third, the risk of cumulative damage may be mathematically linked to duration of exposure, which is logically greater when exposure to concussions begins at a younger age. Lastly, it is important to understand that many cognitive functions are not yet fully developed in pediatric populations, and thus, cognitive assessment tools designed for adults may have significant limitations when applied to young athletes. While mild TBI/concussion, and repeated traumatic injuries associated with sports occur frequently in these younger populations, there is still a dearth of clinical data in grade school and junior high school age children. Considering all these factors related to youth concussion, it is most reasonable to approach the clinical management and return-to-play for these individuals more conservatively than adults, and to call for more translational work targeting both the short and long term effects of repeated mild TBI in the developing brain.

Acknowledgments

This work was supported by: NS27544, NS057420, NS06190, the Child Neurology Foundation/Winokur Family Foundation and the Today’s and Tomorrow’s Children Fund.

Footnotes

The authors have nothing to disclose.

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

References

  • 1.Langlois JA, Rutland-Brown W, Thomas KE. The incidence of traumatic brain injury among children in the United States: differences by race. J Head Trauma Rehabil. 2005;20(3):229–238. doi: 10.1097/00001199-200505000-00006. [DOI] [PubMed] [Google Scholar]
  • 2.Farkas O, Lifshitz J, Povlishock JT. Mechanoporation induced by diffuse traumatic brain injury: an irreversible or reversible response to injury? J Neurosci. 2006 Mar 22;26(12):3130–3140. doi: 10.1523/JNEUROSCI.5119-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Katayama Y, Becker DP, Tamura T, Hovda DA. Massive increases in extracellular potassium and the indiscriminate release of glutamate following concussive brain injury. J Neurosurg. 1990 Dec;73(6):889–900. doi: 10.3171/jns.1990.73.6.0889. [DOI] [PubMed] [Google Scholar]
  • 4.Osteen CL, Giza CC, Hovda DA. Injury-induced alterations in N-methyl-D-aspartate receptor subunit composition contribute to prolonged 45 calcium accumulation following lateral fluid percussion. Neuroscience. 2004;128(2):305–322. doi: 10.1016/j.neuroscience.2004.06.034. [DOI] [PubMed] [Google Scholar]
  • 5.Osteen CL, Moore AH, Prins ML, Hovda DA. Age-dependency of 45calcium accumulation following lateral fluid percussion: acute and delayed patterns. J Neurotrauma. 2001 Feb;18(2):141–162. doi: 10.1089/08977150150502587. [DOI] [PubMed] [Google Scholar]
  • 6.Fineman I, Hovda DA, Smith M, Yoshino A, Becker DP. Concussive brain injury is associated with a prolonged accumulation of calcium: a 45Ca autoradiographic study. Brain Research. 1993;624(1–2):94–102. doi: 10.1016/0006-8993(93)90064-t. [DOI] [PubMed] [Google Scholar]
  • 7.Raghupathi R. Cell death mechanisms following traumatic brain injury. Brain Pathol. 2004 Apr;14(2):215–222. doi: 10.1111/j.1750-3639.2004.tb00056.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sullivan PG, Rabchevsky AG, Waldmeier PC, Springer JE. Mitochondrial permeability transition in CNS trauma: cause or effect of neuronal cell death? J Neurosci Res. 2005 Jan 1–15;79(1–2):231–239. doi: 10.1002/jnr.20292. [DOI] [PubMed] [Google Scholar]
  • 9.Reinert M, Hoelper B, Doppenberg E, Zauner A, Bullock R. Substrate delivery and ionic balance disturbance after severe human head injury. Acta Neurochir Suppl. 2000;76:439–444. doi: 10.1007/978-3-7091-6346-7_91. [DOI] [PubMed] [Google Scholar]
  • 10.Yuen TJ, Browne KD, Iwata A, Smith DH. Sodium channelopathy induced by mild axonal trauma worsens outcome after a repeat injury. J Neurosci Res. 2009 Dec;87(16):3620–3625. doi: 10.1002/jnr.22161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yoshino A, Hovda DA, Kawamata T, Katayama Y, Becker DP. Dynamic changes in local cerebral glucose utilization following cerebral concussion in rats: evidence of a hyper- and subsequent hypometabolic state. Brain Research. 1991;561(1):106–119. doi: 10.1016/0006-8993(91)90755-k. [DOI] [PubMed] [Google Scholar]
  • 12.Kawamata T, Katayama Y, Hovda DA, Yoshino A, Becker DP. Lactate accumulation following concussive brain injury: the role of ionic fluxes induced by excitatory amino acids. Brain Research. 1995;674(2):196–204. doi: 10.1016/0006-8993(94)01444-m. [DOI] [PubMed] [Google Scholar]
  • 13.Kalimo H, Rehncrona S, Soderfeldt B. The role of lactic acidosis in the ischemic nerve cell injury. Acta Neuropathol Suppl (Berl) 1981;7:20–22. doi: 10.1007/978-3-642-81553-9_6. [DOI] [PubMed] [Google Scholar]
  • 14.Glenn TC, Kelly DF, Boscardin WJ, et al. Energy dysfunction as a predictor of outcome after moderate or severe head injury: indices of oxygen, glucose, and lactate metabolism. J Cereb Blood Flow Metab. 2003 Oct;23(10):1239–1250. doi: 10.1097/01.WCB.0000089833.23606.7F. [DOI] [PubMed] [Google Scholar]
  • 15.Martin NA, Patwardhan RV, Alexander MJ, et al. Characterization of cerebral hemodynamic phases following severe head trauma: hypoperfusion, hyperemia, and vasospasm. J Neurosurg. 1997 Jul;87(1):9–19. doi: 10.3171/jns.1997.87.1.0009. [DOI] [PubMed] [Google Scholar]
  • 16.Golding EM, Steenberg ML, Contant CF, Jr, Krishnappa I, Robertson CS, Bryan RM., Jr Cerebrovascular reactivity to CO(2) and hypotension after mild cortical impact injury. Am J Physiol. 1999 Oct;277(4 Pt 2):H1457–1466. doi: 10.1152/ajpheart.1999.277.4.H1457. [DOI] [PubMed] [Google Scholar]
  • 17.Grindel SH. Epidemiology and pathophysiology of minor traumatic brain injury. Curr Sports Med Rep. 2003 Feb;2(1):18–23. doi: 10.1249/00149619-200302000-00005. [DOI] [PubMed] [Google Scholar]
  • 18.Chan KH, Miller JD, Dearden NM. Intracranial blood flow velocity after head injury: relationship to severity of injury, time, neurological status and outcome. J Neurol Neurosurg Psychiatry. 1992 Sep;55(9):787–791. doi: 10.1136/jnnp.55.9.787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mandera M, Larysz D, Wojtacha M. Changes in cerebral hemodynamics assessed by transcranial Doppler ultrasonography in children after head injury. Childs Nerv Syst. 2002 Apr;18(3–4):124–128. doi: 10.1007/s00381-002-0572-5. [DOI] [PubMed] [Google Scholar]
  • 20.Becelewski J, Pierzchala K. Cerebrovascular reactivity in patients with mild head injury. Neurol Neurochir Pol. 2003 Mar-Apr;37(2):339–350. [PubMed] [Google Scholar]
  • 21.Vavilala MS, Lee LA, Boddu K, et al. Cerebral autoregulation in pediatric traumatic brain injury. Pediatr Crit Care Med. 2004;5(3):257–263. doi: 10.1097/01.pcc.0000123545.69133.c3. [DOI] [PubMed] [Google Scholar]
  • 22.Chaiwat O, Sharma D, Udomphorn Y, Armstead WM, Vavilala MS. Cerebral hemodynamic predictors of poor 6-month Glasgow Outcome Score in severe pediatric traumatic brain injury. J Neurotrauma. 2009 May;26(5):657–663. doi: 10.1089/neu.2008.0770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vagnozzi R, Signoretti S, Tavazzi B, et al. Temporal window of metabolic brain vulnerability to concussion: a pilot 1H-magnetic resonance spectroscopic study in concussed athletes--part III. Neurosurgery. 2008 Jun;62(6):1286–1295. doi: 10.1227/01.neu.0000333300.34189.74. discussion 1295–1286. [DOI] [PubMed] [Google Scholar]
  • 24.Tavazzi B, Vagnozzi R, Signoretti S, et al. Temporal window of metabolic brain vulnerability to concussions: oxidative and nitrosative stresses--part II. Neurosurgery. 2007;61(2):390–395. doi: 10.1227/01.neu.0000255525.34956.3f. [DOI] [PubMed] [Google Scholar]
  • 25.Vagnozzi R, Tavazzi B, Signoretti S, et al. Temporal window of metabolic brain vulnerability to concussions: mitochondrial-related impairment--part I. Neurosurgery. 2007;61(2):379–388. doi: 10.1227/01.NEU.0000280002.41696.D8. [DOI] [PubMed] [Google Scholar]
  • 26.Prins ML, Hales A, Reger M, Giza CC, Hovda DA. Repeat traumatic brain injury in the juvenile rat is associated with increased axonal injury and cognitive impairments. Dev Neurosci. 2010;32(5–6):510–518. doi: 10.1159/000316800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Putukian M. Repeat mild traumatic brain injury: how to adjust return to play guidelines. Curr Sports Med Rep. 2006 Feb;5(1):15–22. doi: 10.1097/01.csmr.0000306514.79430.cd. [DOI] [PubMed] [Google Scholar]
  • 28.Kissick J, Johnston KM. Return to play after concussion: principles and practice. Clin J Sport Med. 2005 Nov;15(6):426–431. doi: 10.1097/01.jsm.0000186683.59158.8b. [DOI] [PubMed] [Google Scholar]
  • 29.Guskiewicz KM, McCrea M, Marshall SW, et al. Cumulative effects associated with recurrent concussion in collegiate football players: the NCAA Concussion Study. JAMA. 2003 Nov 19;290(19):2549–2555. doi: 10.1001/jama.290.19.2549. [DOI] [PubMed] [Google Scholar]
  • 30.Saunders RL, Harbaugh RE. The second impact in catastrophic contact-sports head trauma. JAMA. 1984 Jul 27;252(4):538–539. [PubMed] [Google Scholar]
  • 31.Cantu RC. Second-impact syndrome. Clinics in sports medicine. 1998 Jan;17(1):37–44. doi: 10.1016/s0278-5919(05)70059-4. [DOI] [PubMed] [Google Scholar]
  • 32.McCrory PR, Berkovic SF. Second impact syndrome. Neurology. 1998;50(3):677–683. doi: 10.1212/wnl.50.3.677. [DOI] [PubMed] [Google Scholar]
  • 33.Wetjen NM, Pichelmann MA, Atkinson JL. Second impact syndrome: concussion and second injury brain complications. J Am Coll Surg. 2010 Oct;211(4):553–557. doi: 10.1016/j.jamcollsurg.2010.05.020. [DOI] [PubMed] [Google Scholar]
  • 34.Clifton GL, Ziegler MG, Grossman RG. Circulating catecholamines and sympathetic activity after head injury. Neurosurgery. 1981 Jan;8(1):10–14. doi: 10.1227/00006123-198101000-00003. [DOI] [PubMed] [Google Scholar]
  • 35.Lam JM, Hsiang JN, Poon WS. Monitoring of autoregulation using laser Doppler flowmetry in patients with head injury. Journal of Neurosurgery. 1997;86(3):438–445. doi: 10.3171/jns.1997.86.3.0438. [DOI] [PubMed] [Google Scholar]
  • 36.McCrory P. Does second impact syndrome exist? Clin J Sport Med. 2001 Jul;11(3):144–149. doi: 10.1097/00042752-200107000-00004. [DOI] [PubMed] [Google Scholar]
  • 37.Cantu RC, Gean AD. Second-impact syndrome and a small subdural hematoma: an uncommon catastrophic result of repetitive head injury with a characteristic imaging appearance. J Neurotrauma. 2010 Sep;27(9):1557–1564. doi: 10.1089/neu.2010.1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kors EE, Terwindt GM, Vermeulen FL, et al. Delayed cerebral edema and fatal coma after minor head trauma: role of the CACNA1A calcium channel subunit gene and relationship with familial hemiplegic migraine. Ann Neurol. 2001;49(6):753–760. doi: 10.1002/ana.1031. [DOI] [PubMed] [Google Scholar]
  • 39.Longhi L, Saatman KE, Fujimoto S, et al. Temporal window of vulnerability to repetitive experimental concussive brain injury. Neurosurgery. 2005 Feb;56(2):364–374. doi: 10.1227/01.neu.0000149008.73513.44. discussion 364–374. [DOI] [PubMed] [Google Scholar]
  • 40.Doberstein CE, Hovda DA, Becker DP. Clinical considerations in the reduction of secondary brain injury. Ann Emerg Med. 1993 Jun;22(6):993–997. doi: 10.1016/s0196-0644(05)82740-4. [DOI] [PubMed] [Google Scholar]
  • 41.Vagnozzi R, Signoretti S, Tavazzi B, et al. Hypothesis of the postconcussive vulnerable brain: experimental evidence of its metabolic occurrence. Neurosurgery. 2005;57(1):164–171. doi: 10.1227/01.neu.0000163413.90259.85. [DOI] [PubMed] [Google Scholar]
  • 42.Creeley CE, Wozniak DF, Bayly PV, Olney JW, Lewis LM. Multiple episodes of mild traumatic brain injury result in impaired cognitive performance in mice. Acad Emerg Med. 2004 Aug;11(8):809–819. doi: 10.1111/j.1553-2712.2004.tb00761.x. [DOI] [PubMed] [Google Scholar]
  • 43.Garnett MR, Blamire AM, Corkill RG, Cadoux-Hudson TA, Rajagopalan B, Styles P. Early proton magnetic resonance spectroscopy in normal-appearing brain correlates with outcome in patients following traumatic brain injury. Brain. 2000 Oct;123( Pt 10):2046–2054. doi: 10.1093/brain/123.10.2046. [DOI] [PubMed] [Google Scholar]
  • 44.Garnett MR, Blamire AM, Rajagopalan B, Styles P, Cadoux-Hudson TA. Evidence for cellular damage in normal-appearing white matter correlates with injury severity in patients following traumatic brain injury: A magnetic resonance spectroscopy study. Brain. 2000 Jul;123( Pt 7):1403–1409. doi: 10.1093/brain/123.7.1403. [DOI] [PubMed] [Google Scholar]
  • 45.Tran LD, Lifshitz J, Witgen BM, Schwarzbach E, Cohen AS, Grady MS. Response of the contralateral hippocampus to lateral fluid percussion brain injury. J Neurotrauma. 2006 Sep;23(9):1330–1342. doi: 10.1089/neu.2006.23.1330. [DOI] [PubMed] [Google Scholar]
  • 46.Avramescu S, Timofeev I. Synaptic strength modulation after cortical trauma: a role in epileptogenesis. J Neurosci. 2008 Jul 2;28(27):6760–6772. doi: 10.1523/JNEUROSCI.0643-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Salin P, Tseng GF, Hoffman S, Parada I, Prince DA. Axonal sprouting in layer V pyramidal neurons of chronically injured cerebral cortex. J Neurosci. 1995 Dec;15(12):8234–8245. doi: 10.1523/JNEUROSCI.15-12-08234.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Topolnik L, Steriade M, Timofeev I. Hyperexcitability of intact neurons underlies acute development of trauma-related electrographic seizures in cats in vivo. Eur J Neurosci. 2003 Aug;18(3):486–496. doi: 10.1046/j.1460-9568.2003.02742.x. [DOI] [PubMed] [Google Scholar]
  • 49.Dinner DS. Posttraumatic epilepsy. In: La Fibinger., editor. The treatment of epilepsy: principles. Philadelphia; Wyllie E: 1993. pp. 654–658. [Google Scholar]
  • 50.Bergsneider M, Hovda DA, Lee SM, et al. Dissociation of cerebral glucose metabolism and level of consciousness during the period of metabolic depression following human traumatic brain injury. J Neurotrauma. 2000 May;17(5):389–401. doi: 10.1089/neu.2000.17.389. [DOI] [PubMed] [Google Scholar]
  • 51.Fineman I, Hovda DA, Smith M, Yoshino A, Becker DP. Concussive brain injury is associated with a prolonged accumulation of calcium: a 45Ca autoradiographic study. Brain Res. 1993 Oct 8;624(1–2):94–102. doi: 10.1016/0006-8993(93)90064-t. [DOI] [PubMed] [Google Scholar]
  • 52.Ginsberg MD, Zhao W, Alonso OF, Loor-Estades JY, Dietrich WD, Busto R. Uncoupling of local cerebral glucose metabolism and blood flow after acute fluid-percussion injury in rats. The American journal of physiology. 1997 Jun;272(6 Pt 2):H2859–2868. doi: 10.1152/ajpheart.1997.272.6.H2859. [DOI] [PubMed] [Google Scholar]
  • 53.Hovda DA. Metabolic Dysfunction. In: Narayan RK, Wilberger JE, Povlishock JT, editors. Neurotrauma. New York: McGraw-Hill, Inc; 1996. pp. 1459–1478. [Google Scholar]
  • 54.Moore AH, Osteen CL, Chatziioannou AF, Hovda DA, Cherry SR. Quantitative assessment of longitudinal metabolic changes in vivo after traumatic brain injury in the adult rat using FDGmicroPET. J Cereb Blood Flow Metab. 2000 Oct;20(10):1492–1501. doi: 10.1097/00004647-200010000-00011. [DOI] [PubMed] [Google Scholar]
  • 55.Thoenen Neurotrophins and neuronal plasticity. Science. 1995;270(5236):593–598. doi: 10.1126/science.270.5236.593. [DOI] [PubMed] [Google Scholar]
  • 56.Zafra F, Lindholm D, Castren E, Hartikka J, Thoenen H. Regulation of brain-derived neurotrophic factor and nerve growth factor mRNA in primary cultures of hippocampal neurons and astrocytes. J Neurosci. 1992 Dec;12(12):4793–4799. doi: 10.1523/JNEUROSCI.12-12-04793.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tyler WJ, Pozzo-Miller LD. BDNF enhances quantal neurotransmitter release and increases the number of docked vesicles at the active zones of hippocampal excitatory synapses. J Neurosci. 2001 Jun 15;21(12):4249–4258. doi: 10.1523/JNEUROSCI.21-12-04249.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lessmann V, Gottmann K, Heumann R. BDNF and NT–4/5 enhance glutamatergic synaptic transmission in cultured hippocampal neurones. Neuroreport. 1994 Dec 30;6(1):21–25. doi: 10.1097/00001756-199412300-00007. [DOI] [PubMed] [Google Scholar]
  • 59.Levine ES, Dreyfus CF, Black IB, Plummer MR. Brain-derived neurotrophic factor rapidly enhances synaptic transmission in hippocampal neurons via postsynaptic tyrosine kinase receptors. Proc Natl Acad Sci U S A. 1995 Aug 15;92(17):8074–8077. doi: 10.1073/pnas.92.17.8074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Takei N, Sasaoka K, Inoue K, Takahashi M, Endo Y, Hatanaka H. Brain-derived neurotrophic factor increases the stimulation-evoked release of glutamate and the levels of exocytosisassociated proteins in cultured cortical neurons from embryonic rats. J Neurochem. 1997 Jan;68(1):370–375. doi: 10.1046/j.1471-4159.1997.68010370.x. [DOI] [PubMed] [Google Scholar]
  • 61.Neeper SA, Gomez-Pinilla F, Choi J, Cotman C. Exercise and brain neurotrophins. Nature. 1995 Jan 12;373(6510):109. doi: 10.1038/373109a0. [DOI] [PubMed] [Google Scholar]
  • 62.Griesbach GS, Hovda DA, Molteni R, Wu A, Gomez-Pinilla F. Voluntary exercise following traumatic brain injury: brain-derived neurotrophic factor upregulation and recovery of function. Neuroscience. 2004;125(1):129–139. doi: 10.1016/j.neuroscience.2004.01.030. [DOI] [PubMed] [Google Scholar]
  • 63.Ip EY, Zanier ER, Moore AH, Lee SM, Hovda DA. Metabolic, neurochemical, and histologic responses to vibrissa motor cortex stimulation after traumatic brain injury. J Cereb Blood Flow Metab. 2003 Aug;23(8):900–910. doi: 10.1097/01.WCB.0000076702.71231.F2. [DOI] [PubMed] [Google Scholar]
  • 64.Prins ML, Lee SM, Cheng CL, Becker DP, Hovda DA. Fluid percussion brain injury in the developing and adult rat: a comparative study of mortality, morphology, intracranial pressure and mean arterial blood pressure. Brain Res Dev Brain Res. 1996 Sep 2;95(2):272–282. doi: 10.1016/0165-3806(96)00098-3. [DOI] [PubMed] [Google Scholar]
  • 65.Thomas S, Prins ML, Samii M, Hovda DA. Cerebral metabolic response to traumatic brain injury sustained early in development: a 2-deoxy-D-glucose autoradiographic study. Journal of Neurotrauma. 2000;17(8):649–665. doi: 10.1089/089771500415409. [DOI] [PubMed] [Google Scholar]
  • 66.McEwen BS. Stress and hippocampal plasticity. Annu Rev Neurosci. 1999;22:105–122. doi: 10.1146/annurev.neuro.22.1.105. [DOI] [PubMed] [Google Scholar]
  • 67.McEwen BS, Magarinos AM. Stress and hippocampal plasticity: implications for the pathophysiology of affective disorders. Hum Psychopharmacol. 2001 Jan;16(S1):S7–S19. doi: 10.1002/hup.266. [DOI] [PubMed] [Google Scholar]
  • 68.Griesbach GS, Hovda DA, Tio DL, Taylor AN. Heightening of the stress response during the first weeks after a mild traumatic brain injury. Neuroscience. 2011 Mar 31;178:147–158. doi: 10.1016/j.neuroscience.2011.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Avital A, Richter-Levin G. Exposure to juvenile stress exacerbates the behavioural consequences of exposure to stress in the adult rat. Int J Neuropsychopharmacol. 2005 Jun;8(2):163–173. doi: 10.1017/S1461145704004808. [DOI] [PubMed] [Google Scholar]
  • 70.Pryce CR, Ruedi-Bettschen D, Dettling AC, et al. Long-term effects of early-life environmental manipulations in rodents and primates: Potential animal models in depression research. Neurosci Biobehav Rev. 2005;29(4–5):649–674. doi: 10.1016/j.neubiorev.2005.03.011. [DOI] [PubMed] [Google Scholar]
  • 71.Meaney MJ. Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations. Annu Rev Neurosci. 2001;24:1161–1192. doi: 10.1146/annurev.neuro.24.1.1161. [DOI] [PubMed] [Google Scholar]
  • 72.Grundy PL, Harbuz MS, Jessop DS, Lightman SL, Sharples PM. The hypothalamo-pituitary-adrenal axis response to experimental traumatic brain injury. J Neurotrauma. 2001 Dec;18(12):1373–1381. doi: 10.1089/08977150152725669. [DOI] [PubMed] [Google Scholar]
  • 73.Roe SY, McGowan EM, Rothwell NJ. Evidence for the involvement of corticotrophin-releasing hormone in the pathogenesis of traumatic brain injury. Eur J Neurosci. 1998 Feb;10(2):553–559. doi: 10.1046/j.1460-9568.1998.00064.x. [DOI] [PubMed] [Google Scholar]
  • 74.Taylor AN, Rahman SU, Sanders NC, Tio DL, Prolo P, Sutton RL. Injury severity differentially affects short- and long-term neuroendocrine outcomes of traumatic brain injury. J Neurotrauma. 2008 Apr;25(4):311–323. doi: 10.1089/neu.2007.0486. [DOI] [PubMed] [Google Scholar]
  • 75.Shohami E, Bass R, Trembovler V, Weidenfeld J. The effect of the adrenocortical axis upon recovery from closed head injury. J Neurotrauma. 1995 Dec;12(6):1069–1077. doi: 10.1089/neu.1995.12.1069. [DOI] [PubMed] [Google Scholar]
  • 76.Bergsneider M, Hovda DA, McArthur DL, et al. Metabolic recovery following human traumatic brain injury based on FDG-PET: time course and relationship to neurological disability. J Head Trauma Rehabil. 2001;16(2):135–148. doi: 10.1097/00001199-200104000-00004. [DOI] [PubMed] [Google Scholar]
  • 77.McAllister TW, Flashman LA, McDonald BC, Saykin AJ. Mechanisms of working memory dysfunction after mild and moderate TBI: evidence from functional MRI and neurogenetics. J Neurotrauma. 2006 Oct;23(10):1450–1467. doi: 10.1089/neu.2006.23.1450. [DOI] [PubMed] [Google Scholar]
  • 78.Bazarian JJ, Blyth B, Cimpello L. Bench to bedside: evidence for brain injury after concussion--looking beyond the computed tomography scan. Acad Emerg Med. 2006 Feb;13(2):199–214. doi: 10.1197/j.aem.2005.07.031. [DOI] [PubMed] [Google Scholar]
  • 79.Niogi SN, Mukherjee P, Ghajar J, et al. Extent of Microstructural White Matter Injury in Postconcussive Syndrome Correlates with Impaired Cognitive Reaction Time: A 3T Diffusion Tensor Imaging Study of Mild Traumatic Brain Injury. AJNR Am J Neuroradiol. 2008 Feb 26; doi: 10.3174/ajnr.A0970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Johnson JA. The hypothalamic-pituitary-adrenal axis in critical illness. AACN Clin Issues. 2006 Jan-Mar;17(1):39–49. doi: 10.1097/00044067-200601000-00006. [DOI] [PubMed] [Google Scholar]
  • 81.Woolf PD, Cox C, Kelly M, Nichols D, McDonald JV, Hamill RW. The adrenocortical response to brain injury: correlation with the severity of neurologic dysfunction, effects of intoxication, and patient outcome. Alcohol Clin Exp Res. 1990 Dec;14(6):917–921. doi: 10.1111/j.1530-0277.1990.tb01838.x. [DOI] [PubMed] [Google Scholar]
  • 82.Steinbok P, Thompson G. Serum cortisol abnormalities after craniocerebral trauma. Neurosurgery. 1979 Nov;5(5):559–565. doi: 10.1227/00006123-197911000-00003. [DOI] [PubMed] [Google Scholar]
  • 83.Cernak I, Savic VJ, Lazarov A, Joksimovic M, Markovic S. Neuroendocrine responses following graded traumatic brain injury in male adults. Brain Inj. 1999 Dec;13(12):1005–1015. doi: 10.1080/026990599121016. [DOI] [PubMed] [Google Scholar]
  • 84.Cohan P, Wang C, McArthur DL, et al. Acute secondary adrenal insufficiency after traumatic brain injury: a prospective study. Crit Care Med. 2005 Oct;33(10):2358–2366. doi: 10.1097/01.ccm.0000181735.51183.a7. [DOI] [PubMed] [Google Scholar]
  • 85.Bondanelli M, De Marinis L, Ambrosio MR, et al. Occurrence of pituitary dysfunction following traumatic brain injury. J Neurotrauma. 2004 Jun;21(6):685–696. doi: 10.1089/0897715041269713. [DOI] [PubMed] [Google Scholar]
  • 86.Niederland T, Makovi H, Gal V, Andreka B, Abraham CS, Kovacs J. Abnormalities of pituitary function after traumatic brain injury in children. J Neurotrauma. 2007 Jan;24(1):119–127. doi: 10.1089/neu.2005.369ER. [DOI] [PubMed] [Google Scholar]
  • 87.Falkenberg T, Mohammed AK, Henriksson B, Persson H, Winblad B, Lindefors N. Increased expression of brain-derived neurotrophic factor mRNA in rat hippocampus is associated with improved spatial memory and enriched environment. Neuroscience Letters. 1992;138:153–156. doi: 10.1016/0304-3940(92)90494-r. [DOI] [PubMed] [Google Scholar]
  • 88.Kesslak JP, So V, Choi J, Cotman CW, Comez-Pinilla F. Learning upregulates brain-derived neurotrophic factor messenger ribonucleic acid: a mechanism to facilitate encoding and circuit maintenance? Behavioral Neuroscience. 1998;112(4):1012–1019. doi: 10.1037//0735-7044.112.4.1012. [DOI] [PubMed] [Google Scholar]
  • 89.Diamond MC, Lindner B, Raymond A. Extensive cortical depth measurements and neuron size increase in the cortex of environmentally enriched rats. Journal of Comparative Neurology. 1967;131:357–364. [Google Scholar]
  • 90.Rosenzweig MR, Krech D, Bennett EL, Diamond MC. Effects of environmental complexity and training on brain chemistry and anatomy: a replication and extension. J Comp Physiol Psychol. 1962 Aug;55:429–437. doi: 10.1037/h0041137. [DOI] [PubMed] [Google Scholar]
  • 91.Diamond MC, Krech D, Rosenzweig MR. The Effects of an Enriched Environment on the Histology of the Rat Cerebral Cortex. J Comp Neurol. 1964 Aug;123:111–120. doi: 10.1002/cne.901230110. [DOI] [PubMed] [Google Scholar]
  • 92.Fineman I, Giza CC, Nahed BV, Lee SM, Hovda DA. Inhibition of neocortical plasticity during development by a moderate concussive brain injury. J Neurotrauma. 2000 Sep;17(9):739–749. doi: 10.1089/neu.2000.17.739. [DOI] [PubMed] [Google Scholar]
  • 93.Ip EY, Giza CC, Griesbach GS, Hovda DA. Effects of enriched environment and fluid percussion injury on dendritic arborization within the cerebral cortex of the developing rat. J Neurotrauma. 2002 May;19(5):573–585. doi: 10.1089/089771502753754055. [DOI] [PubMed] [Google Scholar]
  • 94.Giza CC, Griesbach GS, Hovda DA. Experience-dependent behavioral plasticity is disturbed following traumatic injury to the immature brain. Behav Brain Res. 2005;157(1):11–22. doi: 10.1016/j.bbr.2004.06.003. [DOI] [PubMed] [Google Scholar]
  • 95.Gurkoff GG, Giza CC, Hovda DA. Lateral fluid percussion injury in the developing rat causes an acute, mild behavioral dysfunction in the absence of significant cell death. Brain Res. 2006 Mar 10;1077(1):24–36. doi: 10.1016/j.brainres.2006.01.011. [DOI] [PubMed] [Google Scholar]
  • 96.Giza CC, Maria NS, Hovda DA. N-methyl-D-aspartate receptor subunit changes after traumatic injury to the developing brain. J Neurotrauma. 2006 Jun;23(6):950–961. doi: 10.1089/neu.2006.23.950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hellmich HL, Garcia JM, Shimamura M, et al. Traumatic brain injury and hemorrhagic hypotension supress neuroprotective gene expression in injured hippocampal neurons. Anesthesiology. 2005;102:806–814. doi: 10.1097/00000542-200504000-00017. [DOI] [PubMed] [Google Scholar]
  • 98.Hicks RR, Li C, Zhang L, Dhillon HS, Prasad MR, Serrogy KB. Alterations in BDNF and trkB mRNA levels in the cerebral cortex following experimental brain trauma in rats. Journal of Neurotrauma. 1999;16(6):501–510. doi: 10.1089/neu.1999.16.501. [DOI] [PubMed] [Google Scholar]
  • 99.Yang K, Perez-Polo, Mu XS, Yan HQ, Xue JJ, Iwamoto Y, Liu SJ, Dixon CE, Hayes RL. Increased expression of brain-derived neurotrophic factor but not neurotrophin-3 mRNA in rat brain after cortical impact injury. Journal of Neuroscience Research. 1996;44(2):157–164. doi: 10.1002/(SICI)1097-4547(19960415)44:2<157::AID-JNR8>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 100.Oyesiku NM, Evans CO, Houston S, Darrell RS, Smith JS, Fulop ZL, Dixon CE, Stein DG. Regional changes in the expression of neurotrophic factors and their receptors following acute traumatic brain injury in the adult rat brain. Brain Research. 1999;833(2):161–172. doi: 10.1016/s0006-8993(99)01501-2. [DOI] [PubMed] [Google Scholar]
  • 101.Schoups AA, Elliott RC, Friedman WJ, Black IB. NGF and BDNF are differentially modulated by visual experience in the developing geniculocortical pathway. Brain Res Dev Brain Res. 1995;86(1–2):326–334. doi: 10.1016/0165-3806(95)00043-d. [DOI] [PubMed] [Google Scholar]
  • 102.Zhou J, Pliego-Rivero B, Bradford HF, Stern GM. The BDNF content of postnatal and adult rat brain: the effects of 6-hydroxydopamine lesions in adult brain. Brain Res Dev Brain Res. 1996 Dec 23;97(2):297–303. doi: 10.1016/s0165-3806(96)00159-9. [DOI] [PubMed] [Google Scholar]
  • 103.Lein ES, Shatz CJ. Rapid regulation of brain-derived neurotrophic factor mRNA within eyespecific circuits during ocular dominance column formation. J Neurosci. 2000 Feb 15;20(4):1470–1483. doi: 10.1523/JNEUROSCI.20-04-01470.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Griesbach GS, Gomez-Pinilla F, Hovda DA. The upregulation of plasticity-related proteins following TBI is disrupted with acute voluntary exercise. Brain Res. 2004 Aug 6;1016(2):154–162. doi: 10.1016/j.brainres.2004.04.079. [DOI] [PubMed] [Google Scholar]
  • 105.Griesbach GS, Hovda DA, Molteni R, Gomez-Pinilla F. Alterations in BDNF and synapsin I within the occipital cortex and hippocampus after mild traumatic brain injury in the developing rat: reflections of injury-induced neuroplasticity. J Neurotrauma. 2002 Jul;19(7):803–814. doi: 10.1089/08977150260190401. [DOI] [PubMed] [Google Scholar]
  • 106.Dancause N, Barbay S, Frost SB, et al. Extensive cortical rewiring after brain injury. J Neurosci. 2005 Nov 2;25(44):10167–10179. doi: 10.1523/JNEUROSCI.3256-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Stein DG, Hoffman SW. Concepts of CNS plasticity in the context of brain damage and repair. J Head Trauma Rehabil. 2003 Jul-Aug;18(4):317–341. doi: 10.1097/00001199-200307000-00004. [DOI] [PubMed] [Google Scholar]
  • 108.Desmurget M, Bonnetblanc F, Duffau H. Contrasting acute and slow-growing lesions: a new door to brain plasticity. Brain. 2007 Apr;130(Pt 4):898–914. doi: 10.1093/brain/awl300. [DOI] [PubMed] [Google Scholar]
  • 109.Dunn-Meynell AA, Levin BE. Lateralized effect of unilateral somatosensory cortex contusion on behavior and cortical reorganization. Brain Res. 1995 Mar 27;675(1–2):143–156. doi: 10.1016/0006-8993(95)00050-z. [DOI] [PubMed] [Google Scholar]
  • 110.Jansen EM, Low WC. Quantitative analysis of contralateral hemisphere hypertrophy and sensorimotor performance in adult rats following unilateral neonatal ischemic-hypoxic brain injury. Brain Res. 1996 Feb 5;708(1–2):93–99. doi: 10.1016/0006-8993(95)01288-5. [DOI] [PubMed] [Google Scholar]
  • 111.Kozlowski D, Schallert T. Relationship between dendritic pruning and behavioral recovery following sensorimotor cortex lesions. Behavioural Brain Research. 1998;97:89–98. doi: 10.1016/s0166-4328(98)00030-8. [DOI] [PubMed] [Google Scholar]
  • 112.Florence SL, Taub HB, Kaas JH. Large-scale sprouting of cortical connections after peripheral injury in adult macaque monkeys. Science. 1998 Nov 6;282(5391):1117–1121. doi: 10.1126/science.282.5391.1117. [DOI] [PubMed] [Google Scholar]
  • 113.Burek MJ, Oppenheim RW. Programmed cell death in the developing nervous system. Brain Pathol. 1996 Oct;6(4):427–446. doi: 10.1111/j.1750-3639.1996.tb00874.x. [DOI] [PubMed] [Google Scholar]
  • 114.Catsicas S, Thanos S, Clarke PG. Major role for neuronal death during brain development: refinement of topographical connections. Proc Natl Acad Sci U S A. 1987 Nov;84(22):8165–8168. doi: 10.1073/pnas.84.22.8165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Adelson PD, Dixon CE, Kochanek PM. Long-term dysfunction following diffuse traumatic brain injury in the immature rat. J Neurotrauma. 2000 Apr;17(4):273–282. doi: 10.1089/neu.2000.17.273. [DOI] [PubMed] [Google Scholar]
  • 116.Anderson V, Catroppa C, Morse S, Haritou F, Rosenfeld J. Functional plasticity or vulnerability after early brain injury? Pediatrics. 2005 Dec;116(6):1374–1382. doi: 10.1542/peds.2004-1728. [DOI] [PubMed] [Google Scholar]
  • 117.Levin HS, Eisenberg HM, Wigg NR, Kobayashi K. Memory and intellectual ability after head injury in children and adolescents. Neurosurgery. 1982 Nov;11(5):668–673. doi: 10.1227/00006123-198211000-00009. [DOI] [PubMed] [Google Scholar]
  • 118.Chapman SB, Levin HS, Wanek A, Weyrauch J, Kufera J. Discourse after closed head injury in young children. Brain Lang. 1998 Feb 15;61(3):420–449. doi: 10.1006/brln.1997.1885. [DOI] [PubMed] [Google Scholar]
  • 119.Klonoff H, Low MD, Clark C. Head injuries in children: a prospective five year follow-up. J Neurol Neurosurg Psychiatry. 1977 Dec;40(12):1211–1219. doi: 10.1136/jnnp.40.12.1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Johnson MH. Sensitive periods in functional brain development: problems and prospects. Dev Psychobiol. 2005 Apr;46(3):287–292. doi: 10.1002/dev.20057. [DOI] [PubMed] [Google Scholar]
  • 121.Giza CC, Prins ML. Is being plastic fantastic? Mechanisms of altered plasticity after developmental traumatic brain injury. Dev Neurosci. 2006;28(4–5):364–379. doi: 10.1159/000094163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Jacobs R, Harvey AS, Anderson V. Executive function following focal frontal lobe lesions: impact of timing of lesion on outcome. Cortex. 2007 Aug;43(6):792–805. doi: 10.1016/s0010-9452(08)70507-0. [DOI] [PubMed] [Google Scholar]
  • 123.Anderson V, Catroppa C. Memory outcome at 5 years post-childhood traumatic brain injury. Brain Inj. 2007 Dec;21(13–14):1399–1409. doi: 10.1080/02699050701785070. [DOI] [PubMed] [Google Scholar]
  • 124.Anderson V, Spencer-Smith M, Coleman L, et al. Children’s executive functions: are they poorer after very early brain insult. Neuropsychologia. 2010 Jun;48(7):2041–2050. doi: 10.1016/j.neuropsychologia.2010.03.025. [DOI] [PubMed] [Google Scholar]
  • 125.Anderson V, Brown S, Newitt H, Hoile H. Long-term outcome from childhood traumatic brain injury: Intellectual ability, personality, and quality of life. Neuropsychology. 2011 Mar;25(2):176–184. doi: 10.1037/a0021217. [DOI] [PubMed] [Google Scholar]
  • 126.Gomez-Pinilla F, Dao L, So V. Physical exercise induces FGF-2 and its mRNA in the hippocampus. Brain Res. 1997 Aug 1;764(1–2):1–8. doi: 10.1016/s0006-8993(97)00375-2. [DOI] [PubMed] [Google Scholar]
  • 127.Carro E, Trejo JL, Busiguina S, Torres-Aleman I. Circulating insulin-like growth factor I mediates the protective effects of physical exercise against brain insults of different etiology and anatomy. J Neurosci. 2001 Aug 1;21(15):5678–5684. doi: 10.1523/JNEUROSCI.21-15-05678.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.van Praag H, Kempermann G, Gage FH. Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci. 1999 Mar;2(3):266–270. doi: 10.1038/6368. [DOI] [PubMed] [Google Scholar]
  • 129.Cotman CW, Berchtold NC. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci. 2002;25(6):295–301. doi: 10.1016/s0166-2236(02)02143-4. [DOI] [PubMed] [Google Scholar]
  • 130.Fabel K, Tam B, Kaufer D, et al. VEGF is necessary for exercise-induced adult hippocampal neurogenesis. Eur J Neurosci. 2003 Nov;18(10):2803–2812. doi: 10.1111/j.1460-9568.2003.03041.x. [DOI] [PubMed] [Google Scholar]
  • 131.van Praag H, Shubert T, Zhao C, Gage FH. Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci. 2005 Sep 21;25(38):8680–8685. doi: 10.1523/JNEUROSCI.1731-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Kleim JA, Jones TA, Schallert T. Motor enrichment and the induction of plasticity before or after brain injury. Neurochem Res. 2003 Nov;28(11):1757–1769. doi: 10.1023/a:1026025408742. [DOI] [PubMed] [Google Scholar]
  • 133.Kolb B. Synaptic plasticity and the organization of behaviour after early and late brain injury. Can J Exp Psychol. 1999 Mar;53(1):62–76. doi: 10.1037/h0087300. [DOI] [PubMed] [Google Scholar]
  • 134.Ivanco TL, Greenough WT. Physiological consequences of morphologically detectable synaptic plasticity: potential uses for examining recovery following damage. Neuropharmacology. 2000 Mar 3;39(5):765–776. doi: 10.1016/s0028-3908(00)00004-6. [DOI] [PubMed] [Google Scholar]
  • 135.Albensi BC. Models of brain injury and alterations in synaptic plasticity. J Neurosci Res. 2001 Aug 15;65(4):279–283. doi: 10.1002/jnr.1151. [DOI] [PubMed] [Google Scholar]
  • 136.Griesbach GS, Hovda DA, Gomez-Pinilla F. Exercise-induced improvement in cognitive performance after traumatic brain injury in rats is dependent on BDNF activation. Brain Res. 2009 Sep 8;1288:105–115. doi: 10.1016/j.brainres.2009.06.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Dishman RK, Berthoud HR, Booth FW, et al. Neurobiology of exercise. Obesity (Silver Spring) 2006 Mar;14(3):345–356. doi: 10.1038/oby.2006.46. [DOI] [PubMed] [Google Scholar]
  • 138.Griesbach GS, Gomez-Pinilla F, Hovda DA. Time window for voluntary exercise-induced increases in hippocampal neuroplasticity molecules after traumatic brain injury is severity dependent. J Neurotrauma. 2007 Jul;24(7):1161–1171. doi: 10.1089/neu.2006.0255. [DOI] [PubMed] [Google Scholar]
  • 139.Smith MA, Makino S, Kvetnansky R, Post RM. Effects of stress on neurotrophic factor expression in the rat brain. Ann NY Acad, Sci. 1995;771:234–239. doi: 10.1111/j.1749-6632.1995.tb44684.x. [DOI] [PubMed] [Google Scholar]
  • 140.Hansson AC, Sommer WH, Metsis M, Stromberg I, Agnati LF, Fuxe K. Corticosterone actions on the hippocampal brain-derived neurotrophic factor expression are mediated by exon IV promoter. J Neuroendocrinol. 2006 Feb;18(2):104–114. doi: 10.1111/j.1365-2826.2005.01390.x. [DOI] [PubMed] [Google Scholar]
  • 141.Gronli J, Bramham C, Murison R, et al. Chronic mild stress inhibits BDNF protein expression and CREB activation in the dentate gyrus but not in the hippocampus proper. Pharmacol Biochem Behav. 2006 Dec;85(4):842–849. doi: 10.1016/j.pbb.2006.11.021. [DOI] [PubMed] [Google Scholar]
  • 142.Hansson AC, Cintra A, Belluardo N, et al. Gluco- and mineralocorticoid receptor-mediated regulation of neurotrophic factor gene expression in the dorsal hippocampus and the neocortex of the rat. Eur J Neurosci. 2000 Aug;12(8):2918–2934. doi: 10.1046/j.1460-9568.2000.00185.x. [DOI] [PubMed] [Google Scholar]
  • 143.Schaaf MJ, De Kloet ER, Vreugdenhil E. Corticosterone effects on BDNF expression in the hippocampus. Implications for memory formation. Stress. 2000 May;3(3):201–208. doi: 10.3109/10253890009001124. [DOI] [PubMed] [Google Scholar]
  • 144.Dishman RK, Renner KJ, White-Welkley JE, Burke KA, Bunnell BN. Treadmill exercise training augments brain norepinephrine response to familiar and novel stress. Brain Res Bull. 2000 Jul 15;52(5):337–342. doi: 10.1016/s0361-9230(00)00271-9. [DOI] [PubMed] [Google Scholar]
  • 145.Dalsgaard MK, Volianitis S, Yoshiga CC, Dawson EA, Secher NH. Cerebral metabolism during upper and lower body exercise. J Appl Physiol. 2004 Nov;97(5):1733–1739. doi: 10.1152/japplphysiol.00450.2004. [DOI] [PubMed] [Google Scholar]
  • 146.Smith PJ, Blumenthal JA, Hoffman BM, et al. Aerobic exercise and neurocognitive performance: a meta-analytic review of randomized controlled trials. Psychosom Med. 2010 Apr;72(3):239–252. doi: 10.1097/PSY.0b013e3181d14633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hillman CH, Erickson KI, Kramer AF. Be smart, exercise your heart: exercise effects on brain and cognition. Nat Rev Neurosci. 2008 Jan;9(1):58–65. doi: 10.1038/nrn2298. [DOI] [PubMed] [Google Scholar]
  • 148.Wipfli BM, Rethorst CD, Landers DM. The anxiolytic effects of exercise: a meta-analysis of randomized trials and dose-response analysis. J Sport Exerc Psychol. 2008 Aug;30(4):392–410. doi: 10.1123/jsep.30.4.392. [DOI] [PubMed] [Google Scholar]
  • 149.Smits JA, Berry AC, Rosenfield D, Powers MB, Behar E, Otto MW. Reducing anxiety sensitivity with exercise. Depress Anxiety. 2008;25(8):689–699. doi: 10.1002/da.20411. [DOI] [PubMed] [Google Scholar]
  • 150.Merom D, Phongsavan P, Wagner R, et al. Promoting walking as an adjunct intervention to group cognitive behavioral therapy for anxiety disorders--a pilot group randomized trial. J Anxiety Disord. 2008 Aug;22(6):959–968. doi: 10.1016/j.janxdis.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 151.Castelli DM, Hillman CH, Buck SM, Erwin HE. Physical fitness and academic achievement in third and fifth-grade students. J Sport Exerc Psychol. 2007 Apr;29(2):239–252. doi: 10.1123/jsep.29.2.239. [DOI] [PubMed] [Google Scholar]
  • 152.Chaddock L, Hillman CH, Buck SM, Cohen NJ. Aerobic fitness and executive control of relational memory in preadolescent children. Med Sci Sports Exerc. 2011 Feb;43(2):344–349. doi: 10.1249/MSS.0b013e3181e9af48. [DOI] [PubMed] [Google Scholar]
  • 153.Hillman CH, Pontifex MB, Raine LB, Castelli DM, Hall EE, Kramer AF. The effect of acute treadmill walking on cognitive control and academic achievement in preadolescent children. Neuroscience. 2009 Mar 31;159(3):1044–1054. doi: 10.1016/j.neuroscience.2009.01.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Davis CL, Tomporowski PD, Boyle CA, et al. Effects of aerobic exercise on overweight children’s cognitive functioning: a randomized controlled trial. Res Q Exerc Sport. 2007 Dec;78(5):510–519. doi: 10.1080/02701367.2007.10599450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Koseoglu E, Akboyraz A, Soyuer A, Ersoy AO. Aerobic exercise and plasma beta endorphin levels in patients with migrainous headache without aura. Cephalalgia. 2003 Dec;23(10):972–976. doi: 10.1046/j.1468-2982.2003.00624.x. [DOI] [PubMed] [Google Scholar]
  • 156.Lockett DM, Campbell JF. The effects of aerobic exercise on migraine. Headache. 1992 Jan;32(1):50–54. doi: 10.1111/j.1526-4610.1992.hed3201050.x. [DOI] [PubMed] [Google Scholar]
  • 157.Gordon WA, Sliwinski M, Echo J, McLoughlin M, Sheerer MS, Meili TE. The benefits of exercise in individuals with traumatic brain injury: a retrospective study. J Head Trauma Rehabil. 1998 Aug;13(4):58–67. doi: 10.1097/00001199-199808000-00006. [DOI] [PubMed] [Google Scholar]
  • 158.Ashman TA, Gordon WA, Cantor JB, Hibbard MR. Neurobehavioral consequences of traumatic brain injury. Mt Sinai J Med. 2006 Nov;73(7):999–1005. [PubMed] [Google Scholar]
  • 159.Bombardier CH, Fann JR, Temkin NR, Esselman PC, Barber J, Dikmen SS. Rates of major depressive disorder and clinical outcomes following traumatic brain injury. JAMA. 2010 May 19;303(19):1938–1945. doi: 10.1001/jama.2010.599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Ekeland E, Heian F, Hagen KB, Abbott J, Nordheim L. Exercise to improve self-esteem in children and young people. Cochrane Database Syst Rev. 2004;(1):CD003683. doi: 10.1002/14651858.CD003683.pub2. [DOI] [PubMed] [Google Scholar]
  • 161.Gagnon I, Galli C, Friedman D, Grilli L, Iverson GL. Active rehabilitation for children who are slow to recover following sport-related concussion. Brain Inj. 2009 Nov;23(12):956–964. doi: 10.3109/02699050903373477. [DOI] [PubMed] [Google Scholar]
  • 162.Majerske CW, Mihalik JP, Ren D, et al. Concussion in sports: postconcussive activity levels, symptoms, and neurocognitive performance. J Athl Train. 2008 May-Jun;43(3):265–274. doi: 10.4085/1062-6050-43.3.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Laurer HL, Bareyre FM, Lee VM, et al. Mild head injury increasing the brain’s vulnerability to a second concussive impact. J Neurosurg. 2001 Nov;95(5):859–870. doi: 10.3171/jns.2001.95.5.0859. [DOI] [PubMed] [Google Scholar]
  • 164.Schulz MR, Marshall SW, Mueller FO, et al. Incidence and risk factors for concussion in high school athletes, North Carolina, 1996–1999. Am J Epidemiol. 2004 Nov 15;160(10):937–944. doi: 10.1093/aje/kwh304. [DOI] [PubMed] [Google Scholar]
  • 165.Aubry M, Cantu R, Dvorak J, et al. Summary and agreement statement of the First International Conference on Concussion in Sport, Vienna 2001. Recommendations for the improvement of safety and health of athletes who may suffer concussive injuries. Br J Sports Med. 2002;36(1):6–10. doi: 10.1136/bjsm.36.1.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Weightman MM, Bolgla R, McCulloch KL, Peterson MD. Physical therapy recommendations for service members with mild traumatic brain injury. J Head Trauma Rehabil. 2010 May-Jun;25(3):206–218. doi: 10.1097/HTR.0b013e3181dc82d3. [DOI] [PubMed] [Google Scholar]
  • 167.McCrory P, Meeuwisse W, Johnston K, et al. Consensus Statement on Concussion in Sport: the 3rd International Conference on Concussion in Sport held in Zurich, November 2008. Br J Sports Med. 2009 May;43( Suppl 1):i76–90. doi: 10.1136/bjsm.2009.058248. [DOI] [PubMed] [Google Scholar]
  • 168.Field AS, Hasan K, Jellison BJ, Arfanakis K, Alexander AL. Diffusion tensor imaging in an infant with traumatic brain swelling. AJNR Am J Neuroradiol. 2003 Aug;24(7):1461–1464. [PMC free article] [PubMed] [Google Scholar]
  • 169.Vagnozzi R, Signoretti S, Cristofori L, et al. Assessment of metabolic brain damage and recovery following mild traumatic brain injury: a multicentre, proton magnetic resonance spectroscopic study in concussed patients. Brain. 2010 Aug 23; doi: 10.1093/brain/awq200. [DOI] [PubMed] [Google Scholar]
  • 170.Pettus EH, Povlishock JT. Characterization of a distinct set of intra-axonal ultrastructural changes associated with traumatically induced alteration in axolemmal permeability. Brain Res. 1996 May 25;722(1–2):1–11. doi: 10.1016/0006-8993(96)00113-8. [DOI] [PubMed] [Google Scholar]
  • 171.Povlishock JT, Christman CW. The pathobiology of traumatically induced axonal injury in animals and humans: a review of current thoughts. Journal of Neurotrauma. 1995;12(4):555–564. doi: 10.1089/neu.1995.12.555. [DOI] [PubMed] [Google Scholar]
  • 172.Buki A, Povlishock JT. All roads lead to disconnection?--Traumatic axonal injury revisited. Acta Neurochir(Wien) 2006;148(2):181–193. doi: 10.1007/s00701-005-0674-4. [DOI] [PubMed] [Google Scholar]
  • 173.Prins ML, Hales A, Reger M, Giza CC, Hovda DA. Repeat Traumatic Brain Injury in the Juvenile Rat Is Associated with Increased Axonal Injury and Cognitive Impairments. Dev Neurosci. 2010 doi: 10.1159/000316800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Reeves TM, Phillips LL, Povlishock JT. Myelinated and unmyelinated axons of the corpus callosum differ in vulnerability and functional recovery following traumatic brain injury. Exp Neurol. 2005;196(1):126–137. doi: 10.1016/j.expneurol.2005.07.014. [DOI] [PubMed] [Google Scholar]
  • 175.Mac Donald CL, Dikranian K, Bayly P, Holtzman D, Brody D. Diffusion tensor imaging reliably detects experimental traumatic axonal injury and indicates approximate time of injury. The Journal of Neuroscience. 2007;27(44):11869–11876. doi: 10.1523/JNEUROSCI.3647-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Mac Donald CL, Dikranian K, Song SK, Bayly PV, Holtzman DM, Brody DL. Detection of traumatic axonal injury with diffusion tensor imaging in a mouse model of traumatic brain injury. Exp Neurol. 2007;205(1):116–131. doi: 10.1016/j.expneurol.2007.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Adams JH, Graham DI, Murray LS, Scott G. Diffuse axonal injury due to nonmissile head injury in humans: an analysis of 45 cases. Ann Neurol. 1982;12(6):557–563. doi: 10.1002/ana.410120610. [DOI] [PubMed] [Google Scholar]
  • 178.Adams JH, Jennett B, Murray LS, Teasdale GM, Gennarelli TA, Graham DI. Neuropathological Findingd in Disabled Survivors of a Head Injury. J Neurotrauma. 2011 Mar 14; doi: 10.1089/neu.2010.1733. [DOI] [PubMed] [Google Scholar]
  • 179.Blumbergs PC, Scott G, Manavis J, Wainwright H, Simpson DA, McLean AJ. Staining of amyloid precursor protein to study axonal damage in mild head injury. Lancet. 1994;344(8929):1055–1056. doi: 10.1016/s0140-6736(94)91712-4. [DOI] [PubMed] [Google Scholar]
  • 180.Blumbergs PC, Scott G, Manavis J, Wainwright H, Simpson DA, McLean AJ. Topography of axonal injury as defined by amyloid precursor protein and the sector scoring method in mild and severe closed head injury. J Neurotrauma. 1995 Aug;12(4):565–572. doi: 10.1089/neu.1995.12.565. [DOI] [PubMed] [Google Scholar]
  • 181.Wozniak JR, Krach L, Ward E, et al. Neurocognitive and neuroimaging correlates of pediatric traumatic brain injury: a diffusion tensor imaging (DTI) study. Arch Clin Neuropsychol. 2007;22(5):555–568. doi: 10.1016/j.acn.2007.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Oni MB, Wilde EA, Bigler ED, et al. Diffusion Tensor Imaging Analysis of Frontal Lobes in Pediatric Traumatic Brain Injury. J Child Neurol. 2010 doi: 10.1177/0883073809356034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Wilde EA, McCauley SR, Hunter JV, et al. Diffusion tensor imaging of acute mild traumatic brain injury in adolescents. Neurology. 2008;70(12):948–955. doi: 10.1212/01.wnl.0000305961.68029.54. [DOI] [PubMed] [Google Scholar]
  • 184.Huang M, Theilmann RJ, Robb A, et al. Integrated imaging approach with MEG and DTI to Detect Mild Traumatic Brain Injury in Military and Civilian Patients. Journal of Neurotrauma. 2009 doi: 10.1089/neu.2008.0672. [DOI] [PubMed] [Google Scholar]
  • 185.Babikian T, Tong KA, Galloway NR, Freier-Randall MC, Obenaus A, Ashwal S. Diffusion-weighted imaging predicts cognition in pediatric brain injury. Pediatr Neurol. 2009;41(6):406–412. doi: 10.1016/j.pediatrneurol.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 186.Niogi SN, Mukherjee P. Diffusion tensor imaging of mild traumatic brain injury. J Head Trauma Rehabil. 2010;25(4):241–255. doi: 10.1097/HTR.0b013e3181e52c2a. [DOI] [PubMed] [Google Scholar]
  • 187.McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. J Neuropathol Exp Neurol. 2009;68(7):709–735. doi: 10.1097/NEN.0b013e3181a9d503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Iwata A, Chen XH, McIntosh TK, Browne KD, Smith DH. Long-term accumulation of amyloid-beta in axons following brain trauma without persistent upregulation of amyloid precursor protein genes. J Neuropathol Exp Neurol. 2002 Dec;61(12):1056–1068. doi: 10.1093/jnen/61.12.1056. [DOI] [PubMed] [Google Scholar]
  • 189.Blasko I, Beer R, Bigl M, et al. Experimental traumatic brain injury in rats stimulates the expression, production and activity of Alzheimer’s disease beta-secretase (BACE-1) J Neural Transm. 2004 Apr;111(4):523–536. doi: 10.1007/s00702-003-0095-6. [DOI] [PubMed] [Google Scholar]
  • 190.Cribbs DH, Chen LS, Cotman CW, LaFerla FM. Injury induces presenilin-1 gene expression in mouse brain. Neuroreport. 1996 Jul 29;7(11):1773–1776. doi: 10.1097/00001756-199607290-00016. [DOI] [PubMed] [Google Scholar]
  • 191.Smith DH, Nakamura M, McIntosh TK, et al. Brain trauma induces massive hippocampal neuron death linked to a surge in beta-amyloid levels in mice overexpressing mutant amyloid precursor protein. Am J Pathol. 1998 Sep;153(3):1005–1010. doi: 10.1016/s0002-9440(10)65643-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Uryu K, Laurer H, McIntosh T, et al. Repetitive mild brain trauma accelerates Abeta deposition, lipid peroxidation, and cognitive impairment in a transgenic mouse model of Alzheimer amyloidosis. J Neurosci. 2002 Jan 15;22(2):446–454. doi: 10.1523/JNEUROSCI.22-02-00446.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Loane DJ, Pocivavsek A, Moussa CE, et al. Amyloid precursor protein secretases as therapeutic targets for traumatic brain injury. Nat Med. 2009 Apr;15(4):377–379. doi: 10.1038/nm.1940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Chen XH, Siman R, Iwata A, Meaney DF, Trojanowski JQ, Smith DH. Long-term accumulation of amyloid-beta, beta-secretase, presenilin-1, and caspase-3 in damaged axons following brain trauma. Am J Pathol. 2004 Aug;165(2):357–371. doi: 10.1016/s0002-9440(10)63303-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Magnoni S, Brody DL. New perspectives on amyloid-beta dynamics after acute brain injury: moving between experimental approaches and studies in the human brain. Arch Neurol. 2010 Sep;67(9):1068–1073. doi: 10.1001/archneurol.2010.214. [DOI] [PubMed] [Google Scholar]
  • 196.Bogdanoff B, Natter HM. Incidence of cavum septum pellucidum in adults: a sign of boxer’s encephalopathy. Neurology. 1989 Jul;39(7):991–992. doi: 10.1212/wnl.39.7.991. [DOI] [PubMed] [Google Scholar]
  • 197.Silbert PL, Gubbay SS, Vaughan RJ. Cavum septum pellucidum and obstructive hydrocephalus. J Neurol Neurosurg Psychiatry. 1993 Jul;56(7):820–822. doi: 10.1136/jnnp.56.7.820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.McKee AC, Gavett BE, Stern RA, et al. TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol. 2010 Sep;69(9):918–929. doi: 10.1097/NEN.0b013e3181ee7d85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Strich SJ. Diffuse degeneration of the cerebral white matter in severe dementia following head injury. J Neurol Neurosurg Psychiatry. 1956 Aug;19(3):163–185. doi: 10.1136/jnnp.19.3.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Gale SD, Burr RB, Bigler ED, Blatter D. Fornix degeneration and memory in traumatic brain injury. Brain Res Bull. 1993;32(4):345–349. doi: 10.1016/0361-9230(93)90198-k. [DOI] [PubMed] [Google Scholar]
  • 201.Pettus EH, Povlishock JT. Characterization of a distinct set of intra-axonal ultrastructural changes associated with traumatically induced alteration in axolemmal permeability. Brain Research. 1996;722(1–2):1–11. doi: 10.1016/0006-8993(96)00113-8. [DOI] [PubMed] [Google Scholar]
  • 202.Binder LI, Guillozet-Bongaarts AL, Garcia-Sierra F, Berry RW. Tau, tangles, and Alzheimer’s disease. Biochim Biophys Acta. 2005 Jan 3;1739(2–3):216–223. doi: 10.1016/j.bbadis.2004.08.014. [DOI] [PubMed] [Google Scholar]
  • 203.Serbest G, Burkhardt MF, Siman R, Raghupathi R, Saatman KE. Temporal profiles of cytoskeletal protein loss following traumatic axonal injury in mice. Neurochem Res. 2007 Dec;32(12):2006–2014. doi: 10.1007/s11064-007-9318-9. [DOI] [PubMed] [Google Scholar]
  • 204.Hof PR, Bouras C, Buee L, Delacourte A, Perl DP, Morrison JH. Differential distribution of neurofibrillary tangles in the cerebral cortex of dementia pugilistica and Alzheimer’s disease cases. Acta Neuropathol. 1992;85(1):23–30. doi: 10.1007/BF00304630. [DOI] [PubMed] [Google Scholar]
  • 205.McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. J Neuropathol Exp Neurol. 2009 Jul;68(7):709–735. doi: 10.1097/NEN.0b013e3181a9d503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Geddes JF, Vowles GH, Nicoll JA, Revesz T. Neuronal cytoskeletal changes are an early consequence of repetitive head injury. Acta Neuropathol. 1999 Aug;98(2):171–178. doi: 10.1007/s004010051066. [DOI] [PubMed] [Google Scholar]
  • 207.Omalu B, Bailes J, Hamilton RL, et al. Emerging Histomorphologic Phenotypes of Chronic Traumatic Encephalopathy [CTE] in American Athletes. Neurosurgery. 2011 Feb 23; doi: 10.1227/NEU.0b013e318212bc7b. [DOI] [PubMed] [Google Scholar]
  • 208.Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem. 2009 May 8;284(19):12845–12852. doi: 10.1074/jbc.M808759200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Johnson VE, Stewart W, Smith DH. Traumatic brain injury and amyloid-beta pathology: a link to Alzheimer’s disease? Nat Rev Neurosci. 2010 Mar 10; doi: 10.1038/nrn2808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Chen XH, Johnson VE, Uryu K, Trojanowski JQ, Smith DH. A lack of amyloid beta plaques despite persistent accumulation of amyloid beta in axons of long-term survivors of traumatic brain injury. Brain Pathol. 2009 Apr;19(2):214–223. doi: 10.1111/j.1750-3639.2008.00176.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Appel SH, Cwik VA, Day JW. Trauma, TDP-43, and amyotrophic lateral sclerosis. Muscle Nerve. 2010 Dec;42(6):851–852. doi: 10.1002/mus.21939. [DOI] [PubMed] [Google Scholar]
  • 212.Armon C, Miller RG. Correspondence regarding: TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol 2010:69;918–29. J Neuropathol Exp Neurol. 2011 Jan;70(1):97–98. doi: 10.1097/01.JNEN.0000392910.86750.32. author reply 98–100. [DOI] [PubMed] [Google Scholar]
  • 213.Zetterberg H, Hietala MA, Jonsson M, et al. Neurochemical aftermath of amateur boxing. Arch Neurol. 2006 Sep;63(9):1277–1280. doi: 10.1001/archneur.63.9.1277. [DOI] [PubMed] [Google Scholar]
  • 214.Zetterberg H, Tanriverdi F, Unluhizarci K, Selcuklu A, Kelestimur F, Blennow K. Sustained release of neuron-specific enolase to serum in amateur boxers. Brain Inj. 2009 Aug;23(9):723–726. doi: 10.1080/02699050903120399. [DOI] [PubMed] [Google Scholar]
  • 215.Saunders AM, Strittmatter WJ, Schmechel D, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology. 1993 Aug;43(8):1467–1472. doi: 10.1212/wnl.43.8.1467. [DOI] [PubMed] [Google Scholar]
  • 216.Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993 Aug 13;261(5123):921–923. doi: 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
  • 217.Strittmatter WJ, Saunders AM, Schmechel D, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A. 1993 Mar 1;90(5):1977–1981. doi: 10.1073/pnas.90.5.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Jordan BD. Chronic traumatic brain injury associated with boxing. Semin Neurol. 2000;20(2):179–185. doi: 10.1055/s-2000-9826. [DOI] [PubMed] [Google Scholar]
  • 219.Jordan BD, Relkin NR, Ravdin LD, Jacobs AR, Bennett A, Gandy S. Apolipoprotein E epsilon4 associated with chronic traumatic brain injury in boxing. JAMA. 1997 Jul 9;278(2):136–140. [PubMed] [Google Scholar]
  • 220.Corder EH, Saunders AM, Risch NJ, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet. 1994 Jun;7(2):180–184. doi: 10.1038/ng0694-180. [DOI] [PubMed] [Google Scholar]

RESOURCES