Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Molecular and Cellular Neuroscience 66 (2015) 81–90

Contents lists available at ScienceDirect

Molecular and Cellular Neuroscience

journal homepage: www.elsevier.com/locate/ymcne

Post-traumatic neurodegeneration and chronic


traumatic encephalopathy
Daniel H. Daneshvar a,b,c, Lee E. Goldstein a,b,c,d,e,f,g,h,i,j, Patrick T. Kiernan a,b,c,
Thor D. Stein a,b,d,k, Ann C. McKee a,b,c,d,k,⁎
a
Boston University Chronic Traumatic Encephalopathy Program, Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
b
Boston University Alzheimer's Disease Center, Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
c
Department of Neurology, Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
d
Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
e
Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
f
Department of Neurosurgery, Boston University School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
g
Boston University Photonics Center, Boston University, 1 Silber Way, Boston, MA 02115, USA
h
Department of Biomedical Engineering, Boston University, 1 Silber Way, Boston, MA 02115, USA
i
Department of Electrical and Computer Engineering, Boston University, 1 Silber Way, Boston, MA 02115, USA
j
Department of Mechanical Engineering, Boston University, 1 Silber Way, Boston, MA 02115, USA
k
VA Boston Healthcare System, 150 South Huntington Avenue, Jamaica Plain, MA 02130, USA

a r t i c l e i n f o a b s t r a c t

Article history: Traumatic brain injury (TBI) is a leading cause of mortality and morbidity around the world. Concussive and
Received 2 February 2015 subconcussive forms of closed-head injury due to impact or blast neurotrauma represent the most common
Accepted 5 March 2015 types of TBI in civilian and military settings. It is becoming increasingly evident that TBI can lead to persistent,
Available online 7 March 2015
long-term debilitating effects, and in some cases, progressive neurodegeneration and chronic traumatic enceph-
alopathy (CTE). The epidemiological literature suggests that a single moderate-to-severe TBI may be associated
Keywords:
Traumatic brain injury
with accelerated neurodegeneration and increased risk of Alzheimer's disease, Parkinson's disease, or motor neu-
Chronic traumatic encephalopathy ron disease. However, the pathologic phenotype of these post-traumatic neurodegenerations is largely unknown
Axonal injury and there may be pathobiological differences between post-traumatic disease and the corresponding sporadic
Brain trauma disorder. By contrast, the pathology of CTE is increasingly well known and is characterized by a distinctive pattern
Posttraumatic neurodegeneration of progressive brain atrophy and accumulation of hyperphosphorylated tau neurofibrillary and glial tangles, dys-
Motor neuron disease trophic neurites, 43 kDa TAR DNA-binding protein (TDP-43) neuronal and glial aggregates, microvasculopathy,
Tau protein myelinated axonopathy, neuroinflammation, and white matter degeneration. Clinically, CTE is associated with
Concussion
behavioral changes, executive dysfunction, memory deficits, and cognitive impairments that begin insidiously
Blast and impact neurotrauma
and most often progress slowly over decades. Although research on the long-term effects of TBI is advancing
quickly, the incidence and prevalence of post-traumatic neurodegeneration and CTE are unknown. Critical
knowledge gaps include elucidation of pathogenic mechanisms, identification of genetic risk factors, and clarifi-
cation of relevant variables—including age at exposure to trauma, history of prior and subsequent head trauma,
substance use, gender, stress, and comorbidities—all of which may contribute to risk profiles and the develop-
ment of post-traumatic neurodegeneration and CTE. This article is part of a Special Issue entitled 'Traumatic
Brain Injury'.
Published by Elsevier Inc.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
1.1. Acute mild TBI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
1.1.1. Concussive and subconcussive injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
1.1.2. Blast injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82

Abbreviations:TDP-43, 43 kDa TAR DNA-binding protein; AD, Alzheimer's disease; APOE ε4, apolipoprotein ε4; Aβ, beta-amyloid; CSF, cerebrospinal fluid; CTE, chronic traumatic enceph-
alopathy; p-tau, hyperphosphorylated tau; mTBI, mild traumatic brain injury; NFTs, neurofibrillary tangles; PHF-tau, paired helical filament-tau; PET, positron emission tomography; PCS,
post-concussion syndrome; TBI, traumatic brain injury.
⁎ Corresponding author at: VA Boston Healthcare System, Boston University School of Medicine, Professor of Neurology and Pathology, Director, CTE Program, Associate Director,
Alzheimer's Disease Center, 150 S. Huntington Avenue, Boston, MA 02130, USA.

http://dx.doi.org/10.1016/j.mcn.2015.03.007
1044-7431/Published by Elsevier Inc.
82 D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90

1.1.3. Juvenile head trauma syndrome and second impact syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83


1.2. Chronic effects of traumatic brain injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
1.2.1. Cognitive deficits and mood disturbances . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
1.2.2. Alzheimer disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
1.2.3. Parkinson disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
1.2.4. Amyotrophic lateral sclerosis or motor neuron disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
2. Chronic traumatic encephalopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.1. Clinical symptoms of CTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.1.1. CTE and PTSD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.2. Neuropathology of chronic traumatic encephalopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.2.1. Gross pathologic features of CTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.2.2. Microscopic pathology of CTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.2.3. TDP-43 pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
2.2.4. Axonal pathology and neuroinflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
2.2.5. Amyloid-β peptide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
2.2.6. Lewy bodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
2.3. Chronic traumatic encephalopathy and co-morbid disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
2.3.1. CTE with motor neuron disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
3. Future areas for research . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
3.1. Potential risk factors for CTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
4. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88

1. Introduction as experimental studies in gyrencephalic piglets demonstrate greater


behavioral abnormalities and more persistent axonal injury in piglets
1.1. Acute mild TBI exposed to sagittal versus axial rotational injury (Sullivan et al., 2013).
The axonal injury produced by mild TBI (mTBI) is multifocal, with a ten-
1.1.1. Concussive and subconcussive injury dency to be most severe in the corpus callosum, fornix, parasagittal
Concussive and subconcussive injuries are thought to be produced white matter and cerebellum, and within these areas, more pronounced
by rapid acceleration and deceleration of the head (Meaney et al., around small blood vessels (McKee et al., 2014).
1995). Rapid linear or angular acceleration, deceleration or rotational These axonal changes likely contribute to the severity of symptoms
forces cause the brain to deform, stretching individual neurons, glial after mTBI and are major contributors to the development of post-
cells and blood vessels and altering membrane permeability. Although concussion syndrome (PCS). Acceleration–deceleration injury also causes
all cell compartments are affected by the injury, blood vessels and tau protein, normally associated with microtubules in axons, to become
axons are especially vulnerable as they often extend long distances abnormally phosphorylated, misfolded, aggregated and cleaved, all of
within the nervous system. In addition to structural deformation, trau- which generate neurotoxic tau peptide fragments (Amadoro et al.,
matic acceleration–deceleration forces produce a rapid influx of calci- 2006; Chen et al., 2010; Kanaan et al., 2011; Khlistunova et al., 2006;
um, efflux of potassium, release of neurotransmitters, and alterations McKee et al., 2013; Zilka et al., 2006). It is not clear how these acute alter-
in the function of cellular sodium–potassium (Na +–K +) pumps. ations develop into a progressive neurodegeneration after repeated injury
These trauma-induced alterations in neuronal homeostasis result in in some individuals, however traumatically-induced microvasculopathy
large increases in glucose metabolism and are collectively referred to with breach of the blood brain barrier and release of normally excluded
as the “neurometabolic cascade of concussion” (Giza and Hovda, 2001, systemic proteins, such as proinflammatory cytokines or other factors
2014). Post-concussive hypermetabolism in the setting of decreased ce- may play a critical role. In addition, recent evidence indicates that a
rebral blood flow produces a disparity between glucose supply and de- brain-wide network of paravascular channels, the “glymphatic” pathway,
mand or a cellular energy crisis (Giza and Hovda, 2001). Pathological facilitates the clearance of interstitial solutes, including tau and beta amy-
studies show that multifocal axonal injury, microhemorrhage, loss of loid (Aβ), from the brain. Experimentally in mice after acute TBI, the
microvascular integrity, and neuroinflammation occur after concussion glymphatic pathway is functionally impaired, an impairment that persists
(Blumbergs et al., 1994; McKee et al., 2014; Oppenheimer, 1968). The for one month post injury and enhances the development of neurofibril-
astrocytosis is most severe in the cerebral white matter and brainstem lary pathology and neurodegeneration in post-traumatic rodent brain.
white matter tracts and clusters of activated microglia are most promi- Chronic impairment of glymphatic pathway function after repetitive TBI
nent in the white matter around small vessels. Perivascular hemosiderin, may be a key factor promoting tau aggregation and the onset of neurode-
hematoidin-laden macrophages and vascular inflammation may also be generation (Iliff et al., 2014).
present after concussion, indicating microvascular damage and breach
of the blood–brain barrier. In addition, focal perivascular accumulations 1.1.2. Blast injury
of hyperphosphorylated tau (p-tau) and hyperphosphorylated TDP-43 Blast injuries resulting from improvised explosive devices have
(p-TDP43) occasionally occur after concussive injury (McKee et al., become an increasingly important form of TBI in civilian and military
2013, 2014). populations. Recent estimates indicate that 10–20% of the 2.5 million
The severity of the axonal injury and microvasculopathy generally U.S. military service members deployed to Iraq and Afghanistan are
parallel the severity of the TBI, with mild injury producing only micro- affected by TBI and the majority of these injuries are associated with
scopic axonal damage and rare microhemorrhages, and moderate to se- blast exposure (The CDC et al., 2013). Individuals exposed to blast are
vere TBI producing more severe axonal injury with grossly visible susceptible to neurological injury with acute and long-term neuropsy-
petechial hemorrhages. The degree of axonal injury after traumatic chiatric and cognitive consequences. Military personnel exposed to re-
impact may also vary with the direction of the head impact rotation, petitive mTBI from explosive blast (Goldstein et al., 2012; McKee and
D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90 83

Robinson, 2014; Omalu et al., 2011) show neuropathological changes of SIS occurs when an individual sustains an initial mild head injury or
early stage CTE, axonopathy, microvascular damage, astrocytosis and concussion, then suffers a second head injury before the symptoms as-
activated microgliosis at autopsy (Goldstein et al., 2012). Clinical symp- sociated with the first injury have cleared, the condition is associated
toms experienced after blast neurotrauma include progressive affective with rapid diffuse cerebral swelling and neurological deterioration
lability, irritability, distractibility, executive dysfunction, memory dis- (Cantu, 1998; Cantu and Gean, 2010; Logan et al., 2001; Miele et al.,
turbances, and cognitive deficits. Four of the five veterans exposed to 2004; Mori et al., 2006; Saunders and Harbaugh, 1984). Typically, the
blast who showed changes of early stage CTE at autopsy were also diag- second injury is only a minor blow to the head, and there is no immedi-
nosed with posttraumatic stress disorder (PTSD) during life suggesting ate loss of consciousness. However, within minutes of the injury, severe
that PTSD and CTE might be biologically and pathologically intercon- cerebrovascular engorgement, cerebral edema and brain herniation de-
nected (McKee and Robinson, 2014). The focal p-tau changes associated velop, associated with precipitous collapse. All reported cases of SIS
with blast neurotrauma share features of early CTE reported in young have involved young athletes, predominantly males (90%) ranging in
American football and soccer players, boxers, head-bangers and others age from 10–24 years, mean age 17.9 years (Mori et al., 2006). Most
(Geddes et al., 1999; Goldstein et al., 2012; McKee et al., 2013, 2014). affected athletes were American football players, usually at the high
However, pathologies associated with blast exposure other than tau ac- school level, but younger players and collegiate athletes have also
cumulation, including axonal injury and microvascular damage, most been reported. SIS has also occurred in association with boxing, karate,
likely are important contributors to the clinical and behavioral abnormal- skiing and ice hockey. It is thought to result from an abrupt posttrau-
ities observed after blast injury. It is worth noting that laboratory mice ex- matic loss of cerebral blood flow autoregulation and catecholamine
posed to a single experimental blast also demonstrate brain pathology, release that produce a rapid increase in intracranial blood volume and
physiologic and functional changes very similar to those found after catastrophic cerebral edema (Clifton et al., 1981; Lam et al., 1997). The
human blast injury—including myelinated axonopathy, focal relationship of SIS to juvenile head trauma syndrome is uncertain, and
microvasculopathy, neuroinflammation, neuronal loss, phosphorylated both may be manifestations of the same underlying pathophysiology.
tau proteinopathy, electrophysiological abnormalities, behavioral impair-
ments, and cognitive deficits (Goldstein et al., 2012). An independent rep- 1.2. Chronic effects of traumatic brain injury
lication study reported brain tau proteinopathy that persisted for at least
one month after exposing mice to a single blast (Huber et al., 2013). A single moderate to severe TBI with loss of consciousness (LOC) is
Blast injuries represent a wide range of heterogeneous injuries and associated with a 2–4 fold increased risk of dementia in later life
are often complicated by other types of TBI, including closed-head (Institute of Medicine Committee on Gulf War and Health: Brain
impact injury (Nakagawa et al., 2011). The occurrence of microscopic Injury in, 2008; Shively et al., 2012). The dementia is most often catego-
neuropathology related to military blast exposure was first reported in rized as probable or possible Alzheimer's disease (AD) using validated
deceased World War I infantry soldiers by Sir Frederick Mott (Mott, clinical criteria, but few studies included neuropathological verification,
1916, 1919). While blast-induced brain pathology has been repeatedly and clinical overlaps between AD, CTE and other post-traumatic
reported in humans and experimental animals, the origins of these inju- neurodegenerations are known to occur (Stern et al., 2013). Studies
ries are only recently beginning to be understood (Goldstein et al., also support a link between a remote single TBI and Parkinson's disease
2014). Kinematic analysis of high-speed videographic records obtained (PD) and amyotrophic lateral sclerosis (ALS), although the pathological
in a military-relevant blast neurotrauma mouse model has shown that features of traumatically induced PD and ALS may differ considerably
blast winds with velocities of more than 330 miles/h—greater than the from the sporadic versions of these neurodegenerations (Daneshvar
most intense wind gust ever recorded on earth—produce rapidly oscil- et al., 2013; Kenney et al., 2014).
lating inertial forces on the head that induce injurious shearing forces
in the brain (Goldstein et al., 2012). An important point is that blast 1.2.1. Cognitive deficits and mood disturbances
winds, not blast waves, are responsible for the resulting cerebral injury, Retired professional football players who sustained 3 or more con-
whereas the acoustic blast wave produces little deformation of brain cussions were found to report more cognitive symptoms including a
tissue as a consequence of rapid shockwave pressure equilibration threefold increase in significant memory impairment, and a fivefold
(Goldstein et al., 2012). Blast injuries may also produce diffuse or focal increase in diagnosed mild cognitive impairment compared to retired
hemorrhage and edema as blood vessels and brain tissue rapidly players without a history of concussion (Guskiewicz et al., 2005).
contract and expand several times within the fraction of a second fol- Other studies found cognitive deficits on neurological and neuropsycho-
lowing transit of the blast shock wave. Some of the traumatic effects logical examination in retired NFL athletes (Ford et al., 2013; Hart et al.,
of blast exposure can be mitigated by immobilizing the head during 2013; Randolph et al., 2013), that were associated with white matter
blast exposure. pathology on DTI and FLAIR imaging and alterations in regional cerebral
blood flow (Hampshire et al., 2013; Hart et al., 2013). A study of fifteen
retired professional football players with a history of concussions and
1.1.3. Juvenile head trauma syndrome and second impact syndrome free of co-morbid conditions showed that there were decreases in frac-
In children and young adults, minor brain trauma can occasionally tional anisotropy and increases in mean and radial diffusivity in the
produce catastrophic, often fatal, cerebral edema and coma. If the neu- frontal–parietal tracts and corpus callosum on DTI compared to an age
rological deterioration occurs after a single TBI, it is referred to as juve- and education matched control group (Tremblay et al., 2014). Further-
nile head trauma syndrome (McQuillen et al., 1988). The neurological more, these changes significantly correlated with a decline in episodic
collapse may be immediate or delayed, occurring after a “lucid interval”. memory decline. Retired NFL players with only mild deficits in execu-
Juvenile head trauma syndrome is thought to represent rapid vasodila- tive functioning showed hyperactivation and hypoconnectivity of the
tion and redistribution of blood into the brain parenchyma after impact dorsolateral frontal and frontopolar cortices on fMRI (Hampshire et al.,
injury, a process that may involve a functional age-related channelopa- 2013). Clinical and functional impairments in cognition have also been
thy. Some individuals with juvenile head trauma syndrome have a mu- correlated with the frequency of head impacts in high school and
tation in the calcium channel subunit gene (CACNA1A) associated with collegiate football players wearing helmet-mounted accelerometers
familial hemiplegic migraine (Kors et al., 2001). Occasionally, juvenile (Breedlove et al., 2012; McAllister et al., 2012; Talavage et al., 2014), al-
head trauma syndrome develops in a young athlete who experiences though not all helmet sensor studies have supported this relationship
two head injuries, with the second injury occurring before complete re- (Broglio et al., 2011; Gysland et al., 2012).
covery from the first impact, similar to second impact syndrome (SIS) Brain trauma experienced in sport has also been linked to distur-
(McQuillen et al., 1988). bances in mood. Retired professional football players who experienced
84 D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90

three or more concussions reported a threefold increase in diagnosed (Schofield et al., 1997). Reanalysis of the Rochester Epidemiology Pro-
depression (Guskiewicz et al., 2007). A follow up survey administered ject data also indicated that the age of onset of AD among AD patients
nine years later provided further evidence for a dose–response relation- with head injury was 8 years earlier than the expected time of onset
ship between self reported concussions and depressive symptoms later predicted by a life-time method based on 689 AD patents without
in life (Kerr et al., 2012). Neuropsychological assessment in former pro- head injury in the same cohort (Guo et al., 2000). Nordstrom et al.
fessional football players has confirmed the relationship between in- evaluated the risk of young onset of dementia (dementia before the
creased self reported concussions and depression (Didehbani et al., age of 65 years) after TBI in a cohort of more than 800,000 young men
2013). Depression in these athletes is also associated with increased followed for over 3 decades. They found a strong dose–response associ-
fractional anisotropy on DTI, as well as white matter abnormalities on ation between TBI and young onset dementia, with more severe or more
structural imaging (Hart et al., 2013; Strain et al., 2013). frequent TBI associated with increasing risk (Nordström et al., 2014).
Most case control and longitudinal epidemiologic studies of demen-
1.2.2. Alzheimer disease tia after TBI use only clinical criteria to diagnose AD. Recent clinical
In a meta-analysis of 15 case–control studies, males who had a single studies show that CTE and other post-traumatic, neuropathologically
head injury associated with loss of consciousness (LOC) had a 50% in- verified degenerations share considerable clinical features with AD
creased risk of AD dementia (Fleminger et al., 2003). In a recent study and may be incorrectly diagnosed as probable AD. There have been
of older veterans, a history of TBI was associated with a 60% increase only a few reports of head injury that used neuropathological confirma-
in the risk of developing dementia and an accelerated age of onset by tion of the AD diagnosis. Jelling and colleagues examined the incidence
2 years (Barnes et al., 2014). In the MIRAGE study, head injury increased of AD pathology in 55 consecutive autopsy cases with a history of single
the risk of AD and the magnitude of the risk was proportional to the se- TBI. They found definite or probable AD in 21.8% that was significantly
verity of the head trauma and heightened among first-degree relatives higher than the 14% expected in an age-matched general population
of AD patients (Guo et al., 2000). A longitudinal study involving World (Jellinger et al., 2001).
War II Navy veterans also demonstrated increased risk for AD in The pathological link between Alzheimer's neurodegeneration and
late life with increasing TBI severity (Plassman et al., 2000). Veterans TBI may be related to the accumulation of Aβ and tau proteins after
with severe TBI with loss of consciousness (LOC) or prolonged post- trauma. Aβ plaques and intra-axonal Aβ deposits have been found
traumatic amnesia (PTA) were 4 times more likely to have AD, whereas in approximately one third of TBI subjects dying shortly after injury
veterans with moderate TBI were twice as likely to have AD in late-life (DeKosky et al., 2007; Gentleman et al., 1997; Ikonomovic et al.,
compared to controls (Plassman et al., 2000). No increased risk was 2004), even in young subjects (Horsburgh et al., 2000), and may contin-
found for veterans who had a mTBI (LOC or PTA less than 30 min). ue to accumulate chronically (Johnson et al., 2010). Murine models also
Other studies indicate that while moderate–severe TBI increases the show transient elevation of amyloid precursor protein (APP), and intra-
risk for dementia age groups older than 55 years, mTBI increases the axonal Aβ deposits after acute TBI (Chen et al., 2004). Neuropathological
risk of dementia only among individuals older than 65 years (Gardner analysis of 18 cases of fatal TBI showed widespread axonal injury, accu-
et al., 2014). mulation of neurofilament protein, amyloid APP, Aβ and alpha-
Other studies have also suggested that TBI precipitates an earlier synuclein in axonal bulbs and varicosities (Uryu et al., 2007). p-Tau pro-
onset of AD. In the Northern Manhattan study, a history of head injury tein was also found to accumulate in both axons and neuronal cell bod-
with LOC was associated with earlier onset of AD dementia, whereas ies (Uryu et al., 2007). p-Tau immunoreactive neurofibrillary tangles
mTBI was not significantly associated with earlier onset of AD dementia, (NFTs) have been observed in young individuals dying weeks to months
and those with severe TBI were at significantly increased risk for AD after their last concussion (McKee et al., 2014). An autopsy study of 32

Fig. 1. Atypical AD-like neurodegeneration, Lewy body disease, and axonopathy in a 62 year old woman 11 years after a single severe TBI.
D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90 85

long-term single TBI survivors (age range: 19–60 years, mean 48 years, symptoms may result from neuronal loss in the substantia nigra associ-
93% male, survival 1–47 years, mean 8.4 years after TBI) found wide- ated with either accumulation of alpha-synuclein in Lewy bodies, as oc-
spread Aβ plaques and p-tau NFTs in one third of subjects. NFTs were curs in PD, or p-tau inclusions in the form of NFTs, as occurs in CTE, AD
particularly increased in subjects under the age of 39 years compared and many other tauopathies, it is likely that non-alpha synuclein pathol-
to age-matched controls. The NFTs were described as superficial and ogies contribute to the increased frequency of Parkinsonism following
slightly clustered at sulcal depths suggesting that a single, moderate to TBI (Uryu et al., 2003).
severe traumatic injury may promote the development of an atypical
AD-like neurodegeneration (Johnson et al., 2010). In an individual
case report, a 62 year old woman developed progressive dementia 1.2.4. Amyotrophic lateral sclerosis or motor neuron disease
and parkinsonism 11 years after a severe motor vehicle accident; at ALS is a fatal progressive degeneration of motor neurons in the brain
autopsy, her brain showed multiple neurodegenerative processes and spinal cord. Ninety to 95% of ALS cases are sporadic; gene mutations
including atypical AD, Lewy body disease, axonopathy and TDP-43 in copper/zinc superoxide dismutase 1, senataxin, dynactin, angiogenic,
proteinopathy (Fig. 1). A strikingly similar case was reported by Kenney and TAR-DBP (the gene for TDP-43 on chromosome 1) account for some
and colleagues (Kenney et al., 2014). The subject was a 75-year-old man familial forms of the disease (Bruijn et al., 2004). ALS is pathologically
who developed a rapidly progressive dementia 12 years before death characterized by motor neuron loss and corticospinal tract degenera-
and 25 years after a moderate to severe TBI. Neuropathological exami- tion. Remaining motor neurons in sporadic ALS often have ubiquitin-
nation revealed atypical AD with hippocampal sparing and severe and TDP-43 immunoreactive inclusion bodies that appear either as
white matter degeneration. These reports suggest that moderate–se- rounded or skein-like inclusions.
vere TBI may generate unique neurodegenerative processes with clini- Although the etiology of sporadic ALS is unknown, it may involve in-
copathological phenotypes unlike sporadic disease (Kenney et al., teraction between genetic and environmental risk factors. Many envi-
2014). It is also worth noting that many individuals who suffer TBI do ronmental risk factors have been considered as possible triggers of
not develop dementia, as suggested by Gardner and Yaffe “post-TBI neu- ALS neurodegeneration, including a history of trauma to the brain and
rodegeneration is a multifactorial process that is likely dependent upon spinal cord (Chen et al., 2007; Schmidt et al., 2010; Strickland et al.,
number, mechanics, and timing of TBIs, individual genetics, and many 1996). Recent literature points toward a trend not only between CNS
other health-related, lifestyle, and environmental risk and protective trauma and the development of ALS but also between a smaller number
factors” (Gardner and Yaffe, 2014). of years between the last injury and ALS diagnosis, and older age at the
At autopsy, her brain showed multiple neurodegenerative processes last injury and the development of ALS (Schmidt et al., 2010). In a case
including widespread diffuse plaques in the neocortex (A, B (Aβ immu- control study of 109 cases of ALS and 255 controls, Chen et al. (2007)
nostaining), C (Bielschowsky silver method)), Lewy body disease with found that having experienced repeated head injuries or having been
abnormally large Lewy bodies in the thalamus and mammillary bodies injured within the 10 years before diagnosis was associated with a
(D, E, F (alpha-synuclein immunostaining)), axonopathy with extreme- more than 3-fold higher risk of ALS, with a slightly elevated risk for
ly large axonal spheroids in the thalamus (G (SMI-34 immunostain), H the interval 11 to 30 years. The authors also further performed a
(AT8 immunostaining)), and an atypical distribution of p-tau NFTS meta-analysis of 8 previous ALS studies and estimated a pooled OR of
and dystrophic neurites (AT8 immunostaining) (I, locus coeruleus, J, K, 1.7 (95% CI, 1.3–2.2) for at least one previous head injury. Another
temporal cortex, L, median raphe), all calibration bars = 100 μm. case–control study, which was not included in the meta-analysis, re-
Several investigators have studied the relationship between inheri- ported an increased risk of ALS when the last head injury occurred at
tance of an apolipoprotein ε4 (APOE ε4) allele and dementia after TBI. an older age and closer to the time of diagnosis (Binazzi et al., 2009).
In the Northern Manhattan population study, a history of TBI and inher- ALS incidence and mortality are also reported to be unusually high
itance of an APOE ε4 allele were associated with a 10-fold increased risk among professional soccer players in Italy (Chio et al., 2005). ALS mor-
of AD, while APOE ε4 in the absence of TBI resulted in only a 2-fold tality for Italian professional soccer players was increased 12-fold,
increased risk (Mayeux et al., 1995). These results are in contrast to whereas mortality from other causes was generally lower or compara-
findings in the MIRAGE study in which head injury increased the odds ble to that of the general population (Belli and Vanacore, 2005). Further-
of AD to a greater extent among those lacking the APOE ε4 allele com- more, an incidence study involving 7325 Italian professional soccer
pared to those having one or two ε4 alleles (Guo et al., 2000). players showed that the incidence of ALS was 6.5 times higher than ex-
There is also evidence that APOE ε4 is associated with deposition of Aβ pected (Chio et al., 2005). A study looking at cause of death in retired
protein after head injury (Hartman et al., 2002). APOE ε4 has been associ- NFL players who played for 5 years or more were found have a 4.31
ated with increased severity of chronic neurologic deficits in boxers with higher risk of developing ALS and a 3.86 higher risk of developing de-
high exposure to repetitive trauma compared to those without an APOE mentia compared to age and gender matched controls. Although the
ε4 allele (Jordan et al., 1997). In addition, inheritance of an APOE ε4 allele death certificates indicated dementia and ALS, there was no neuropath-
is associated with longer periods of unconsciousness following severe ological verification of the neurodegenerative processes, and the actual
blunt traumatic injury as well as poorer functional outcome (Friedman underlying diagnoses might well have included CTE and CTE with motor
et al., 1999). Furthermore, Aβ deposition in CTE is associated with the neuron disease (CTE-MND) (Lehman et al., 2012).
APOE ε4 allele and significantly increased in CTE compared to a normal
aging population (Thor Stein, personal communication).

1.2.3. Parkinson disease


In addition to AD, a link has been suggested between TBI and PD. In a
Table 1
case–control study, data from the Rochester Epidemiology Project was
Characteristics of the hyperphosphorylated tau pathology in CTE.
used to identify 196 subjects who developed PD and compared to con- Adapted from McKee et al., Brain 2013.
trols matched for age and gender. The frequency of head trauma was
1. Perivascular foci of p-tau immunoreactive astrocytic tangles (ATs) and
significantly higher in PD cases compared to controls (OR 4.3). The OR
neurofibrillary tangles (NFTs)
for PD was substantially increased (11.0) in subjects who experienced 2. Irregular cortical distribution of p-tau immunoreactive NFTs and ATs with a
mTBI with LOC or more severe TBI (Bower et al., 2003). Animal studies predilection for the depth of cerebral sulci
have shown that the brains of aged TBI-injured mice develop a transient 3. Clusters of subpial and periventricular ATs in the cerebral cortex, diencephalon,
increase in alpha-synuclein that is not found in sham-injured aged ani- and brainstem
4. NFTs in the cerebral cortex located preferentially in the superficial layers
mals or TBI-injured young mice (Uryu et al., 2006). As parkinsonian
86 D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90

2. Chronic traumatic encephalopathy 2.1.1. CTE and PTSD


The clinical course of CTE (and some TBI) frequently overlaps with
2.1. Clinical symptoms of CTE posttraumatic stress disorder (PTSD). All 3 conditions are characterized
by neuropsychiatric symptoms indicative of frontal lobe dysfunction
The symptoms of CTE are insidious, often first manifested as dis- including alterations in working memory, planning, multitasking, com-
turbances in attention or concentration or depression that are occa- plex decision making, judgment, empathy, executive function, impulsivi-
sionally associated with headaches. Short-term memory difficulties, ty, emotional lability, and disinhibition, as well as changes in personality
aggressive tendencies, executive dysfunction and explosivity are and social behavior. Some individuals diagnosed with PTSD have neuro-
also frequent symptoms. Characteristically the first symptoms usual- pathological changes of CTE at autopsy (Goldstein et al., 2012), and it is
ly appear around ages 35–45 years, although the range is broad, from possible that CTE, axonal injury and neuroinflammation may underlie
24 years to 65 years (McKee et al., 2013). There is characteristically a some of the symptoms that are associated with PTSD.
long latent period (mean 8 years, range 0–37 years) between the last
trauma and the development of symptoms (McKee et al., 2009), 2.2. Neuropathology of chronic traumatic encephalopathy
however some athletes develop symptoms while playing that do
not improve once sports participation has stopped. In these individ- Like many neurodegenerative diseases, CTE can only be diagnosed
uals, it is unclear if the symptoms represent prolonged post- definitively at post-mortem neuropathological examination (McKee
concussive symptoms or the early manifestations of CTE. Unlike et al., 2009, 2013; Stern et al., 2013) and no consensus criteria or bio-
other neurodegenerative disorders, headache is a persistent and markers of disease currently exist that can be used to aid in the clinical
early symptom in nearly half the individuals who develop CTE. The diagnosis of CTE (Stern et al., 2013). Corsellis, Bruton, and Freeman-
headache may be migrainous or a constant dull pain. Browne described the clinical symptoms and neuropathological find-
Other personality and behavioral changes that are common in ings in 15 former boxers (Corsellis et al., 1973). They noted a reduction
individuals with early (Stage I or II) CTE are irritability, explosivity, in brain weight, enlargement of the lateral and third ventricles, thinning
and erratic behaviors (McKee et al., 2013). Early-stage CTE is often of the corpus callosum, cavum septum pellucidum with fenestrations,
asymptomatic, and the symptoms that may be present, including head- and scarring and neuronal loss of the cerebellar tonsils. Using histolog-
ache, loss of attention and concentration, depression and irritability are ical stains including silver stains they also described neurofibrillary
non-specific (Daneshvar et al., 2011b). Subjects with Stage II CTE are degeneration of the substantia nigra and cerebral cortex, often in the ab-
usually symptomatic and demonstrate short-term memory difficulties, sence of senile plaques. Subsequent studies using immunocytochemical
aggressive tendencies, difficulties with planning and organization, techniques determined that many of the original cases from Corsellis
mood swings and explosivity (McKee et al., 2013; Stern et al., 2013). showed widespread diffuse Aβ deposits. Recent analysis over 140
By Stage III disease, most subjects are considered cognitively impaired cases of CTE from the VA-BU-SLI CTE Brain Bank determined that 67%
and by Stage IV CTE, executive dysfunction and memory loss are usually of CTE cases have no evidence of Aβ deposition either as diffuse or neu-
severe and most are demented. Other symptoms often include im- ritic senile plaques or amyloid angiopathy. Furthermore, Aβ deposition
pulsivity, suicidality, language difficulties, paranoia, and visuospatial in this large series was significantly associated with age at death, and no
abnormalities; approximately one third are suicidal at some point in subjects less than 50 years demonstrated Aβ deposition as diffuse or
their course. Gait disturbances, dysarthria, and Parkinsonism are neuritic plaques.
found in about 10% of individuals with CTE in association with late
stage disease. 2.2.1. Gross pathologic features of CTE
Recent case series of athletes with neuropathologically confirmed The most commonly observed gross pathologic features of CTE are
CTE suggested that there are three distinct presentations of CTE: one generalized cerebral atrophy, with a tendency to affect the frontal, tem-
with a younger age at onset that presents as behavioral disturbances poral and medial temporal lobes most severely, thalamic, hypothalamic
(e.g., impulsivity, violence) or mood changes (e.g., depression, hopeless- and mammillary body atrophy, enlargement of the lateral and third
ness), another that manifests later in life as cognitive impairment ventricles, thinning of the corpus callosum, cavum septum pellucidum,
(e.g., episodic memory deficits, executive dysfunction), and a mixed septal fenestrations, and pallor of the substantia nigra and locus
presentation that presents with both behavioral, mood and cognitive coeruleus (Daneshvar et al., 2011b; McKee et al., 2009, 2010). Cerebellar
symptoms (Montenigro et al., 2014; Stern et al., 2013). Demented sub- scarring, a feature recorded often in early reports of boxers, is not a com-
jects with neuropathologically diagnosed with Stage IV CTE often meet mon feature of CTE in modern day athletes, although there is often mi-
clinical criteria for AD established by the National Institute on Aging- croscopic evidence of p-tau neurodegeneration in the dentate nucleus
Alzheimer's Association (NIA-AA) Workgroup and the NINCDS-ADRDA of the cerebellum and roof of the fourth ventricle.
(Stern et al., 2013). Current research is focused on delineating aspects of
the clinical presentation that could help distinguish CTE from AD, in addi- 2.2.2. Microscopic pathology of CTE
tion to the history of neurotrauma (Montenigro et al., 2014).
In CTE, the severity of cognitive impairment is most likely associated 2.2.2.1. Hyperphosphorylated tau (Table 1). CTE is a tauopathy, and mi-
with several pathologies, including neuroinflammation, axonal, neuro- croscopically there are widespread deposits of p-tau protein as NFTs
nal and synapse loss and the accumulation of toxic aggregates of tau throughout the brain (McKee et al., 2009, 2013). Tau in CTE is similar
and TDP proteins. The severity of the tau pathology almost certainly biochemically to tau in AD. It is composed of both 3- and 4-
contributes to cognitive decline and behavioral changes, as has been microtubule binding repeat tau, with a ratio of 4 versus 3 microtubule
shown for AD (McKee et al., 1991), however, there is also substantial binding repeat tau of ~ 1 (although the pattern of neuronal 4R tau in
axonal pathology and neuroinflammation in CTE that might contribute CTE differs from AD) and six abnormally phosphorylated tau isoforms
to the development of personality and behavior changes, especially in (McKee et al., 2013; Stern et al., 2013). Two microscopic features unique
early stages of disease when the accumulations of p-tau are minimal. to CTE, not found in another tauopathy, are that the tau pathology in CTE
Other contributors to clinical symptoms include accumulations of is 1) perivascular 2) irregularly distributed in the depths of cortical sulci.
toxic forms of pTDP-43. Although the clinical symptoms of CTE often The early p-tau deposition of CTE affects the cerebral cortex, primarily
begin with behavior and personality changes in a person's thirties or the frontal, temporal, septal and insular neocortex, although the ento-
forties similar to frontotemporal lobar degeneration (FTLD), the rhinal and parahippocampal cortex may occasionally be affected in
clinical course of CTE is considerably slower and often spans three young subjects. Unlike the early stages of AD, however, the hippocam-
or four decades. pus is spared in early phases of CTE. Abnormal p-tau astrocytes are
D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90 87

also commonly found in the subpial region at the sulcal depths. There 2.2.4. Axonal pathology and neuroinflammation
may be scattered tau deposition in the ependyma lining the ventricles In addition to p-tau pathology, axonal pathology is also present at all
and on the lateral surface of the brainstem. Scattered tau positive axonal stages of CTE and progresses with CTE severity. In the earliest stages of
varicosities and neuropil threads are found in the subcortical and deep disease, distorted axonal varicosities are found in the cortex, subcortical
white matter. white matter, and deep white matter tracts of the diencephalon. By
Stage III CTE, severe axonal loss and pathological profiles are found in
2.2.2.2. Stages of CTE: evolution of p-tau pathology. the subcortical white matter, and are most severe in the frontal and
temporal lobes. In advanced CTE, there is widespread axonal loss with
Stage I: In Stage I CTE, tau is found in focal, perivascular clusters as frequent severely distorted axonal profiles widely distributed in the
NFTs and dystrophic neurites, often at the sulcal depths of subcortical white matter (McKee et al., 2013).
frontal, septal or temporal cortex. The surrounding cortex is In addition to degeneration of myelinated axons, neuroinflamma-
typically unremarkable although rare NFTs may be found in tion is a consistent feature of CTE, perivascular clusters of activated mi-
the deep nuclei, such as the locus coeruleus. croglia are found throughout the subcortical white matter in early stage
Stage II: In Stage II CTE, multiple discrete clusters of perivascular p-tau CTE, in later stage disease, dense activated microglia are found through-
NFTs are found in the sulcal depths, most commonly in fron- out the gray and white matter. A robust astrocytosis also found in the
tal, temporal, parietal, insular and septal cortices. NFTs are white matter of early stages of CTE, in more advanced disease, generalized
also found in the superficial layers of adjacent cortex sur- astrocytosis is found throughout the gray and white matter.
rounding the epicenters. The nucleus basalis of Meynert
and locus coeruleus show NFTs as well. Rare NFTs may be 2.2.5. Amyloid-β peptide
found in entorhinal cortex, amygdala, hippocampus, thala- Aβ containing plaques, especially diffuse Aβ plaques, are present in
mus, substantia nigra, and dorsal and median raphe nuclei some cases of CTE, although they are not a consistent feature of CTE,
of the midbrain. are not found in early stage disease, and are significantly associated
Stage III: In Stage III CTE, the medial temporal lobe structures including with age at death (McKee et al., 2013). Compared to a normal aging
the hippocampus, entorhinal cortex, and amygdala show neu- population, Aβ deposition occurs at an earlier age and at an accelerated
rofibrillary degeneration, and there is widespread involvement rate in CTE, and is associated with increased clinical and pathological se-
of the frontal, temporal parietal, insula and septal cortices. verity (Thor Stein, personal communication).
Moderate densities of NFTs are found in olfactory bulbs, hypo-
thalamus, thalamus, mammillary bodies, substantia nigra and 2.2.6. Lewy bodies
dorsal and median raphe nuclei. Alpha-synuclein positive Lewy bodies are found in approximately
Stage IV: In Stage IV CTE, there are dense NFTs in widespread regions of 20% of CTE cases, and are significantly associated with the age of the
the brain, with prominent neuronal loss and gliosis of the neo- subject at death (McKee et al., 2013).
cortex and increasing p-tau in astrocytes. There is prominent
neuronal loss in CA1 of the hippocampus and subiculum, cere- 2.3. Chronic traumatic encephalopathy and co-morbid disease
bral cortices and increasing myelinated fiber and axonal loss in
the white matter. Severe p-tau pathology is found in the cere- Like most other neurodegenerative diseases (Gorell et al., 1994;
bral cortex, diencephalon, basal ganglia, and brainstem, white Jicha and Carr, 2010; Kalaria, 2000; Sonnen et al., 2007; Waite et al.,
matter tracts and spinal cord. Primary visual cortex is spared. 1997), CTE is associated with the development of other neuro-
degenerations, especially as a function of increased age (Hazrati et al.,
2013; McKee et al., 2010, 2013). Of 142 neuropathologically confirmed
2.2.3. TDP-43 pathology cases of CTE, 15 (10.56%) had coexistent motor neuron disease (MND), 15
The majority of CTE cases also demonstrate TDP-43 immunoreactive (10.56%) AD, 10 (7.04%) Lewy Body disease, 4 (2.82%) frontotemporal
intraneuronal and intraglial inclusions and neurites (McKee et al., lobar degeneration (FTLD), and 7 (4.93%) had two or more of these
2010). The pathology ranges from cytoplasmic neuronal inclusions comorbidities; 89 (63.16%) cases were diagnosed only with CTE.
that partially co-localize with p-tau inclusions to glial inclusions and
abnormal neurites (McKee et al., 2010). TDP-43 immunoreactivity 2.3.1. CTE with motor neuron disease
increases with the degree of tau pathology, and is found in nearly all Approximately 10–13% of individuals with CTE develop a progres-
Stage IV cases as TDP-43 positive rounded and threadlike neurites, sive motor neuron disease (MND) that is clinically indistinguishable
intraglial and intraneuronal inclusions. Cases with the most severe from sporadic ALS (McKee et al., 2010, 2013). Typically, individuals
TDP-43 deposition show dense accumulations of TDP-43 inclusions with CTE and MND present with motor weakness, atrophy and fascicu-
and neurites in all layers of the neocortex, particularly layer II, as well lations and develop mild cognitive and behavioral symptoms several
as occasional TDP-43-positive inclusions in the dentate fascia of the hip- years after the onset of their motor disorder. Among the 142 neuro-
pocampus, a distribution pattern that overlaps with the distribution pathologically confirmed cases of CTE, there were 16 individuals who de-
of TDP-43 found in frontotemporal lobar degeneration with TDP-43 veloped a progressive motor neuron disease (MND) (11.4%) (McKee et al.,
(FTLD-TDP) (Cairns et al., 2007). 2013, 2014). Most subjects presented with motor weakness, atrophy and
TDP-43 is an RNA-binding protein that regulates RNA metabolism fasciculations and developed mild cognitive and behavioral symptoms
(Sephton et al., 2010, 2012) and binds to tau (Sato et al., 2009). Dysreg- several years after the onset of motor symptoms. The mechanisms under-
ulation of TDP-43 may influence tau isoform expression and the devel- lying the development of MND after trauma remain unknown but p-tau
opment of tau pathology (Morales et al., 2009). Diseases caused by lesions are frequently found in early CTE, and electrophysiological abnor-
abnormal TDP-43 metabolism have shown altered tau metabolism malities have been found in the motor cortex after concussion in young
including hyperphosphorylation, tau phosphatase resistance, and depo- athletes (Pearce et al., 2014) in retired athletes (De Beaumont et al.,
sition of p-tau intracellular aggregates (Strong and Yang, 2011). In ex- 2013). Furthermore, there is recent evidence suggesting that lesions to
perimental in mouse models of TBI, either from controlled cortical the motor cortex induce amyotrophic lateral sclerosis (Rosenbohm
impact or blast injury, elevated breakdown fragments of TDP-43 are et al., 2014). Multiple experimental models of TBI in mice have demon-
found (Yang et al., 2014). In addition, TDP-43 and its 35 kDa fragment strated increased fragmentation of TDP-43 and relocation of TDP-43
levels are elevated in the cerebrospinal fluid (CSF) of patients with from its normal position in the nucleus to the cytoplasm where it is asso-
severe TBI. ciated with degeneration (Moisse et al., 2009a,b; Yang et al., 2014).
88 D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90

3. Future areas for research Cognitive reserve and cognitive flexibility are thought to play a
significant role in the development and clinical course of CTE. In other
Other than trauma, the risk factors for CTE remain unknown, but are neurodegenerative diseases such as AD, cognitive reserve is considered
likely multifactorial. Genotypic, diagnostic, and prognostic markers are protective against clinical manifestations (Stern et al., 2013) and that
urgently needed to assess CTE risk, resilience, and responsivity to treat- one can increase one's “reserve” and relative resistance with occupa-
ment in individuals at risk for the disease. Other factors such as gender, tional and educational attainment (Stern et al., 2013). In diseases such
age at first exposure, number, timing and severity of head injuries, as AD and CTE, cognitive reserve provides an explanation for individual
cognitive reserve and flexibility, substance use and neuropsychiatric co- differences in measured cognitive deficits despite similar degrees of
morbidities are likely modulators of CTE diathesis and disease progres- neuropathology (Stern et al., 2013).
sion. The contribution of these and other factors to CTE pathogenesis are
poorly understood, especially in the context of individual patients. At 4. Conclusions
present, CTE is a postmortem diagnosis. Consequently, clinicians are in
critical need of objective diagnostic biomarkers to enable reliable detec- TBI, particularly mTBI, has been largely overlooked as a major health
tion, staging, and tracking of CTE during life. Biomarkers are also needed concern until very recently. It is increasingly clear that TBI is a process
for assessment of presymptomatic, incipient, and prodromal disease. and not a static injury, and that prolonged symptoms in TBI survivors
Development of sensitive and specific diagnostics is essential for identi- represent functional and structural damage. Moderate-to-severe TBI
fication of individuals “at risk” who may benefit from early intervention may be associated with accelerated neurodegeneration and increased
as emerging therapeutics become available. Moreover, reliable bio- risk of Alzheimer's disease, Parkinson's disease, and motor neuron dis-
markers for CTE are required to monitor therapeutic and rehabilitative ease, although it is if the pathologic phenotype of these post-traumatic
efficacy, as well as to guide comprehensive multidisciplinary care neurodegenerations differs from sporadic disease. While repetitive
services for patients and their families. Understanding the mechanistic mild TBI is associated with the development of a tauopathy, CTE, very
underpinnings of CTE pathobiology is an essential prerequisite for de- little is known about the pathogenetic mechanisms underlying the de-
velopment of new treatments, diagnostics, and rehabilitative strategies velopment of a neurodegeneration after the acute insults. Effective
for this devastating disease. For this there is no substitute for “bench-to- treatment of mTBI, concussion and subconcussion will need to address
bedside” translational research (Goldstein et al., 2014). the diagnosis and management of the acute injury as well as the long-
term progressive neurodegeneration that occasionally follow. Currently,
3.1. Potential risk factors for CTE the best therapy is prevention and continued public education about
proper detection and management of the acute traumatic injuries.
Several studies indicate that the APOE ε4 allele may be associated
with a poorer prognosis in patients with severe TBI (Zeng et al., 2014).
Allelic variation in APOE in boxers and professional football players Acknowledgments
has also been implicated in prolonged recovery time and poorer cogni-
tive outcome after a single TBI (Jordan et al., 1997). Several studies have We gratefully acknowledge the use of resources and facilities at
reported a higher than expected prevalence of APOE ε4 carriers and the Edith Nourse Rogers Memorial Veterans Hospital (Bedford,
homozygotes in individuals with neuropathologically confirmed CTE MA). We also gratefully acknowledge the help of all members of
(McKee et al., 2009, 2013; Stern et al., 2013). While no genes have the Boston University and the Boston VA, and the individuals and
been established as risk factors for CTE, these findings suggest that families whose participation and contributions made this work
APOE ε4 may increase risk of developing CTE. Triple transgenic mice possible. This work was supported by the Department of Veterans
crossed with the APOE ε4 genotype had more axonal injury after TBI Affairs; Veterans Affairs Biorepository (CSP 501); Translational
compared to triple transgenic mice crossed with APOE ε2 or APOE ε3, al- Research Center for Traumatic Brain Injury and Stress Disorders
though there was no significant effect on Aβ or p-tau deposition (TRACTS) Veterans Affairs Rehabilitation Research and Develop-
(Bennett et al., 2013). Other candidate risk factor genes include MAPT ment Traumatic Brain Injury Center of Excellence (B6796-C);
and TARDBP (Czell et al., 2013; Lee et al., 2013). National Institute of Neurological Diseases and Stroke 1U01NS086659-
The discovery of highly-penetrant tau gene (MAPT) mutations in 01, National Institute of Aging Boston University Alzheimer's Disease
rare families with FTLD demonstrated that tau dysfunction alone was Center [P30AG13846; supplement 0572063345-5]; National Institute of
sufficient to cause neurodegeneration (Gasparini et al., 2007). Some Aging Boston University Framingham Heart Study R01 [AG1649]; Sports
MAPT mutations, clustered around exon 10, influence splicing, which Legacy Institute; and National Operating Committee on Standards for
leads to accumulation of tau having four microtubule binding domain Athletic Equipment. This work was also supported by an unrestricted
repeats (4R) over those with three (3R) (Vandrovcova et al., 2010). gift from the National Football League, the Andlinger Foundation and
MAPT also harbors a common genetic variation that is associated with the WWE.
tauopathy. MAPT is contained within a ~900 kb ancestral genomic inver-
sion that defines two haplotypes, H1 and H2 (Stefansson et al., 2005). References
These haplotypes are in complete linkage disequilibrium and do not re-
combine. Sporadic tauopathies such as progressive supranuclear palsy Amadoro, G., Ciotti, M.T., Costanzi, M., Cestari, V., Calissano, P., Canu, N., 2006. NMDA re-
ceptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation.
and corticobasal degeneration as well as PD are associated with the H1 Proc. Natl. Acad. Sci. U. S. A. 103 (8), 2892–2897. http://dx.doi.org/10.1073/pnas.
haplotype (Baker et al., 1999; Bekris et al., 2010; Di Maria et al., 2000). 0511065103.
It remains to be determined if the H1 haplotype pays a role in susceptibil- Baker, M., Litvan, I., Houlden, H., Adamson, J., Dickson, D., Perez-Tur, J., . . ., Hutton, M.,
1999. Association of an extended haplotype in the tau gene with progressive
ity for CTE. supranuclear palsy. Hum. Mol. Genet. 8 (4), 711–715.
Gender differences may play a role in the development of CTE. To Barnes, D.E., Kaup, A., Kirby, K.A., Byers, A.L., Diaz-Arrastia, R., Yaffe, K., 2014. Traumatic
date, most neuropathologically confirmed cases have been identified brain injury and risk of dementia in older veterans. Neurology 83 (4), 312–319.
Bekris, L.M., Mata, I.F., Zabetian, C.P., 2010. The genetics of Parkinson disease. J. Geriatr.
in men, largely reflecting the demographic of TBI. CTE has been docu-
Psychiatry Neurol. 23 (4), 228–242.
mented in 2 women (reviewed in McKee et al., 2009), it if women are Belli, S., Vanacore, N., 2005. Proportionate mortality of Italian soccer players: is amyotro-
as susceptible to CTE as men. Women appear to be at greater risk for phic lateral sclerosis an occupational disease? Eur. J. Epidemiol. 20 (3), 237–242.
mTBI and PCS-related symptoms (Daneshvar et al., 2011b). These differ- Bennett, R.E., Esparza, T.J., Lewis, H.A., Kim, E., Mac Donald, C.L., Sullivan, P.M., Brody, D.L.,
2013. Human apolipoprotein E4 worsens acute axonal pathology but not amyloid-β
ences may based on hormonal differences, neck and muscle strength, or immunoreactivity after traumatic brain injury in 3xTG-AD mice. J. Neuropathol.
simply more reporting of somatic symptoms (Daneshvar et al., 2011a). Exp. Neurol. 72 (5), 396–403.
D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90 89

Binazzi, A., Belli, S., Uccelli, R., Desiato, M.T., Talamanca, I.F., Antonini, G., . . ., Vanacore, N., Gentleman, S.M., Greenberg, B.D., Savage, M.J., Noori, M., Newman, S.J., Roberts,
2009. An exploratory case–control study on spinal and bulbar forms of amyotrophic G.W., . . ., Graham, D.I., 1997. A β42 is the predominant form of amyloid b‐pro-
lateral sclerosis in the province of Rome. Amyotroph. Lateral Scler. 10 (5–6), tein in the brains of short‐term survivors of head injury. Neuroreport 8 (6),
361–369. http://dx.doi.org/10.3109/17482960802382313. 1519–1522.
Blumbergs, P.C., Scott, G., Manavis, J., Wainwright, H., Simpson, D.A., McLean, A., 1994. Giza, C.C., Hovda, D.A., 2001. The neurometabolic cascade of concussion. J. Athl. Train. 36
Staining of amyloid precursor protein to study axonal damage in mild head injury. (3), 228–235.
Lancet 344 (8929), 1055–1056. Giza, C.C., Hovda, D.A., 2014. The new neurometabolic cascade of concussion. Neurosurgery
Bower, J.H., Maraganore, D.M., Peterson, B.J., McDonnell, S.K., Ahlskog, J.E., Rocca, W.A., 2003. 75 (Suppl. 4), S24–S33. http://dx.doi.org/10.1227/neu.0000000000000505.
Head trauma preceding PD: a case–control study. Neurology 60 (10), 1610–1615. Goldstein, L.E., Fisher, A.M., Tagge, C.A., Zhang, X.L., Velisek, L., Sullivan, J.A., . . ., McKee,
Breedlove, E.L., Robinson, M., Talavage, T.M., Morigaki, K.E., Yoruk, U., O'Keefe, K., . . ., Nauman, A.C., 2012. Chronic traumatic encephalopathy in blast-exposed military veterans
E.A., 2012. Biomechanical correlates of symptomatic and asymptomatic neurophysiolog- and a blast neurotrauma mouse model. Sci. Transl. Med. 4 (134), 1–16. http://dx.
ical impairment in high school football. J. Biomech. 45 (7), 1265–1272. doi.org/10.1126/scitranslmed.3003716.
Broglio, S.P., Eckner, J.T., Surma, T., Kutcher, J.S., 2011. Post-concussion cognitive declines Goldstein, L.E., McKee, A.C., Stanton, P.K., 2014. Considerations for animal models of blast-
and symptomatology are not related to concussion biomechanics in high school foot- related traumatic brain injury and chronic traumatic encephalopathy. Alzheimers
ball players. J. Neurotrauma 28 (10), 2061–2068. Res. Ther. 6 (5), 64. http://dx.doi.org/10.1186/s13195-014-0064-3.
Bruijn, L.I., Miller, T.M., Cleveland, D.W., 2004. Unraveling the mechanisms involved in Gorell, J.M., Johnson, C.C., Rybicki, B.A., 1994. Parkinson's disease and its comorbid
motor neuron degeneration in ALS. Annu. Rev. Neurosci. 27, 723–749. http://dx.doi. disorders: an analysis of Michigan mortality data, 1970 to 1990. Neurology 44 (10),
org/10.1146/annurev.neuro.27.070203.144244. 1865–1868.
Cairns, N.J., Neumann, M., Bigio, E.H., Holm, I.E., Troost, D., Hatanpaa, K.J., . . ., Kretzschmar, Guo, Z., Cupples, L.A., Kurz, A., Auerbach, S.H., Volicer, L., Chui, H., . . ., Farrer, L.A., 2000.
H.A., 2007. TDP-43 in familial and sporadic frontotemporal lobar degeneration with Head injury and the risk of AD in the MIRAGE study. Neurology 54 (6), 1316–1323.
ubiquitin inclusions. Am. J. Pathol. 171 (1), 227–240. Guskiewicz, K.M., Marshall, S.W., Bailes, J., McCrea, M., Cantu, R.C., Randolph, C., Jordan,
Cantu, R.C., 1998. Second-impact syndrome. Clin. Sports Med. 17 (1), 37–44. http://dx.doi. B.D., 2005. Association between recurrent concussion and late-life cognitive impairment
org/10.1016/S0278-5919(05)70059-4. in retired professional football players. Neurosurgery 57 (4), 719–726.
Cantu, R.C., Gean, A.D., 2010. Second-impact syndrome and a small subdural hematoma: Guskiewicz, K.M., Marshall, S.W., Bailes, J., McCrea, M., Harding, H.P., Matthews, A., . . .,
an uncommon catastrophic result of repetitive head injury with a characteristic im- Cantu, R.C., 2007. Recurrent concussion and risk of depression in retired professional
aging appearance. J. Neurotrauma 27 (9), 1557–1564. football players. Med. Sci. Sports Exerc. 39 (6), 903.
Chen, X.H., Siman, R., Iwata, A., Meaney, D.F., Trojanowski, J.Q., Smith, D.H., 2004. Long- Gysland, S.M., Mihalik, J.P., Register-Mihalik, J.K., Trulock, S.C., Shields, E.W., Guskiewicz,
term accumulation of amyloid-beta, beta-secretase, presenilin-1, and caspase-3 in K.M., 2012. The relationship between subconcussive impacts and concussion history
damaged axons following brain trauma. Am. J. Pathol. 165 (2), 357–371. on clinical measures of neurologic function in collegiate football players. Ann.
Chen, H., Richard, M., Sandler, D.P., Umbach, D.M., Kamel, F., 2007. Head injury and Biomed. Eng. 40 (1), 14–22.
amyotrophic lateral sclerosis. Am. J. Epidemiol. 166 (7), 810–816. http://dx.doi.org/ Hampshire, A., MacDonald, A., Owen, A.M., 2013. Hypoconnectivity and hyperfrontality in
10.1093/aje/kwm153. retired American football players. Sci. Rep. 3.
Chen, L.J., Wang, Y.J., Tseng, G.F., 2010. Compression alters kinase and phosphatase activ- Hart, J., Kraut, M.A., Womack, K.B., Strain, J., Didehbani, N., Bartz, E., . . ., Cullum, C.M., 2013.
ity and tau and MAP2 phosphorylation transiently while inducing the fast adaptive Neuroimaging of cognitive dysfunction and depression in aging retired National Foot-
dendritic remodeling of underlying cortical neurons. J. Neurotrauma 27 (9), ball League players: a cross-sectional study. JAMA Neurol. 70 (3), 326–335.
1657–1669. http://dx.doi.org/10.1089/neu.2010.1308. Hartman, R.E., Laurer, H., Longhi, L., Bales, K.R., Paul, S.M., McIntosh, T.K., Holtzman, D.M.,
Chio, A., Benzi, G., Dossena, M., Mutani, R., Mora, G., 2005. Severely increased risk of 2002. Apolipoprotein E4 influences amyloid deposition but not cell loss after trau-
amyotrophic lateral sclerosis among Italian professional football players. Brain 128 matic brain injury in a mouse model of Alzheimer's disease. J. Neurosci. 22 (23),
(Pt 3), 472–476. http://dx.doi.org/10.1093/brain/awh373. 10083–10087.
Clifton, G.L., Ziegler, M.G., Grossman, R.G., 1981. Circulating catecholamines and sympa- Hazrati, L.-N., Tartaglia, M.C., Diamandis, P., Davis, K.D., Green, R.E., Wennberg, R., . . .,
thetic activity after head injury. Neurosurgery 8 (1), 10–14. Tator, C.H., 2013. Absence of chronic traumatic encephalopathy in retired football
Corsellis, J., Bruton, C., Freeman-Browne, D., 1973. The aftermath of boxing. Psychol. Med. players with multiple concussions and neurological symptomatology. Front. Hum.
3 (03), 270–303. Neurosci. 7.
Czell, D., Andersen, P.M., Morita, M., Neuwirth, C., Perren, F., Weber, M., 2013. Phenotypes Horsburgh, K., Cole, G.M., Yang, F., Savage, M.J., Greenberg, B.D., Gentleman, S.M., . . ., Nicoll,
in Swiss patients with familial ALS carrying TARDBP mutations. Neurodegener. Dis. J.A., 2000. Beta-amyloid (Abeta)42(43), abeta42, abeta40 and apoE immunostaining of
12 (3), 150–155. http://dx.doi.org/10.1159/000345835. plaques in fatal head injury. Neuropathol. Appl. Neurobiol. 26 (2), 124–132.
Daneshvar, D.H., Nowinski, C.J., McKee, A.C., Cantu, R.C., 2011a. The epidemiology of Huber, B.R., Meabon, J.S., Martin, T.J., Mourad, P.D., Bennett, R., Kraemer, B.C., . . ., Cook,
sport-related concussion. Clin. Sports Med. 30 (1), 1–17. D.G., 2013. Blast exposure causes early and persistent aberrant phospho- and
Daneshvar, D.H., Riley, D.O., Nowinski, C.J., McKee, A.C., Stern, R.A., Cantu, R.C., 2011b. cleaved-tau expression in a murine model of mild blast-induced traumatic brain injury.
Long-term consequences: effects on normal development profile after concussion. J. Alzheimers Dis. 37 (2), 309–323. http://dx.doi.org/10.3233/jad-130182.
Phys. Med. Rehabil. Clin. N. Am. 22 (4), 683–700. Ikonomovic, M.D., Uryu, K., Abrahamson, E.E., Ciallella, J.R., Trojanowski, J.Q., Lee, V.M., . . .,
Daneshvar, D., Katz, D., Stein, T., Alvarez, V., McKee, A., 2013. Alzheimer's disease, Lewy DeKosky, S.T., 2004. Alzheimer's pathology in human temporal cortex surgically
body disease and TDP43 proteinopathy following a single TBI. Alzheimers Dement. excised after severe brain injury. Exp. Neurol. 190 (1), 192–203. http://dx.doi.org/
9 (4), P438. http://dx.doi.org/10.1016/j.jalz.2013.05.868. 10.1016/j.expneurol.2004.06.011.
De Beaumont, L., Tremblay, S., Henry, L.C., Poirier, J., Lassonde, M., Théoret, H., 2013. Motor Iliff, J.J., Chen, M.J., Plog, B.A., Zeppenfeld, D.M., Soltero, M., Yang, L., . . ., Nedergaard, M.,
system alterations in retired former athletes: the role of aging and concussion history. 2014. Impairment of glymphatic pathway function promotes tau pathology after
BMC Neurol. 13 (1), 109. traumatic brain injury. J. Neurosci. 34 (49), 16180–16193.
DeKosky, S.T., Abrahamson, E.E., Ciallella, J.R., Paljug, W.R., Wisniewski, S.R., Clark, R.S., Institute of Medicine Committee on Gulf War and Health: Brain Injury in, V. a. L.-T. H.,
Ikonomovic, M.D., 2007. Association of increased cortical soluble abeta42 levels Outcomes, 2008. Gulf War and Health: Volume 7: Long-term Consequences of Traumatic
with diffuse plaques after severe brain injury in humans. Arch. Neurol. 64 (4), Brain Injury. National Academies Press (US), Washington (DC).
541–544. http://dx.doi.org/10.1001/archneur.64.4.541. Jellinger, K.A., Paulus, W., Wrocklage, C., Litvan, I., 2001. Traumatic brain injury as a risk
Di Maria, E., Tabaton, M., Vigo, T., Abbruzzese, G., Bellone, E., Donati, C., . . ., Mandich, P., factor for Alzheimer disease. Comparison of two retrospective autopsy cohorts with
2000. Corticobasal degeneration shares a common genetic background with progres- evaluation of ApoE genotype. BMC Neurol. 1 (1), 3.
sive supranuclear palsy. Ann. Neurol. 47 (3), 374–377. Jicha, G.A., Carr, S.A., 2010. Conceptual evolution in Alzheimer's disease: implications for
Didehbani, N., Cullum, C.M., Mansinghani, S., Conover, H., Hart, J., 2013. Depressive symp- understanding the clinical phenotype of progressive neurodegenerative disease.
toms and concussions in aging retired NFL players. Arch. Clin. Neuropsychol. 28 (5), J. Alzheimers Dis. 19 (1), 253–272. http://dx.doi.org/10.3233/jad-2010-1237.
418–424. Johnson, V.E., Stewart, W., Smith, D.H., 2010. Traumatic brain injury and amyloid-beta
Fleminger, S., Oliver, D., Lovestone, S., Rabe-Hesketh, S., Giora, A., 2003. Head injury as a pathology: a link to Alzheimer's disease? Nat. Rev. Neurosci. 11 (5), 361–370. http://
risk factor for Alzheimer's disease: the evidence 10 years on; a partial replication. dx.doi.org/10.1038/nrn2808.
J. Neurol. Neurosurg. Psychiatry 74 (7), 857–862. Jordan, B.D., Relkin, N.R., Ravdin, L.D., Jacobs, A.R., Bennett, A., Gandy, S., 1997. Apolipoprotein
Ford, J.H., Giovanello, K.S., Guskiewicz, K.M., 2013. Episodic memory in former professional E epsilon4 associated with chronic traumatic brain injury in boxing. JAMA 278 (2),
football players with a history of concussion: an event-related functional neuroimaging 136–140.
study. J. Neurotrauma 30 (20), 1683–1701. Kalaria, R.N., 2000. The role of cerebral ischemia in Alzheimer's disease. Neurobiol. Aging
Friedman, G., Froom, P., Sazbon, L., Grinblatt, I., Shochina, M., Tsenter, J., . . ., Groswasser, Z., 21 (2), 321–330.
1999. Apolipoprotein E-epsilon4 genotype predicts a poor outcome in survivors of Kanaan, N.M., Morfini, G.A., LaPointe, N.E., Pigino, G.F., Patterson, K.R., Song, Y., . . ., Binder,
traumatic brain injury. Neurology 52 (2), 244–248. L.I., 2011. Pathogenic forms of tau inhibit kinesin-dependent axonal transport
Gardner, R.C., Yaffe, K., 2014. Traumatic brain injury may increase risk of young onset through a mechanism involving activation of axonal phosphotransferases. J. Neurosci.
dementia. Ann. Neurol. 75 (3), 339–341. 31 (27), 9858–9868.
Gardner, R.C., Burke, J.F., Nettiksimmons, J., Kaup, A., Barnes, D.E., Yaffe, K., 2014. Dementia Kenney, K., Massetti, S., Shively, S., Diaz-Arrastia, R., Perl, D., 2014. Clinicopatho-
risk after traumatic brain injury vs nonbrain trauma: the role of age and severity. logical correlation of a case of dementia after TBI. Alzheimers Dement. 10 (4),
JAMA Neurol. 71 (12), 1490–1497. P843.
Gasparini, L., Terni, B., Spillantini, M.G., 2007. Frontotemporal dementia with tau pathology. Kerr, Z.Y., Marshall, S.W., Harding, H.P., Guskiewicz, K.M., 2012. Nine-year risk of depres-
Neurodegener. Dis. 4 (2–3), 236–253. sion diagnosis increases with increasing self-reported concussions in retired profes-
Geddes, J., Vowles, G., Nicoll, J., Revesz, T., 1999. Neuronal cytoskeletal changes are sional football players. Am. J. Sports Med. 40 (10), 2206–2212.
an early consequence of repetitive head injury. Acta Neuropathol. 98 (2), Khlistunova, I., Biernat, J., Wang, Y., Pickhardt, M., von Bergen, M., Gazova, Z., . . .,
171–178. Mandelkow, E.-M., 2006. Inducible expression of Tau repeat domain in cell models
90 D.H. Daneshvar et al. / Molecular and Cellular Neuroscience 66 (2015) 81–90

of tauopathy aggregation is toxic to cells but can be reversed by inhibitor drugs. Randolph, C., Karantzoulis, S., Guskiewicz, K., 2013. Prevalence and characterization of
J. Biol. Chem. 281 (2), 1205–1214. mild cognitive impairment in retired national football league players. J. Int.
Kors, E.E., Terwindt, G.M., Vermeulen, F.L., Fitzsimons, R.B., Jardine, P.E., Heywood, P., . . ., Neuropsychol. Soc. 19 (08), 873–880.
Frants, R.R., 2001. Delayed cerebral edema and fatal coma after minor head trauma: Rosenbohm, A., Kassubek, J., Weydt, P., Marroquin, N., Volk, A.E., Kubisch, C., . . .,
role of the CACNA1A calcium channel subunit gene and relationship with familial Weishaupt, J.H., 2014. Can lesions to the motor cortex induce amyotrophic lateral
hemiplegic migraine. Ann. Neurol. 49 (6), 753–760. sclerosis? J. Neurol. 261 (2), 283–290.
Lam, J.M., Hsiang, J.N., Poon, W.S., 1997. Monitoring of autoregulation using laser Sato, T., Takeuchi, S., Saito, A., Ding, W., Bamba, H., Matsuura, H., . . ., Urushitani, M., 2009.
Doppler flowmetry in patients with head injury. J. Neurosurg. 86 (3), 438–445. Axonal ligation induces transient redistribution of TDP-43 in brainstem motor neu-
Lee, S.E., Tartaglia, M.C., Yener, G., Genc, S., Seeley, W.W., Sanchez-Juan, P., . . ., Miller, B.L., rons. Neuroscience 164 (4), 1565–1578.
2013. Neurodegenerative disease phenotypes in carriers of MAPT p.A152T, a risk Saunders, R.L., Harbaugh, R.E., 1984. The second impact in catastrophic contact-sports
factor for frontotemporal dementia spectrum disorders and Alzheimer disease. head trauma. JAMA 252 (4), 538–539.
Alzheimer Dis. Assoc. Disord. 27 (4), 302–309. http://dx.doi.org/10.1097/WAD. Schmidt, S., Kwee, L.C., Allen, K.D., Oddone, E.Z., 2010. Association of ALS with head injury,
0b013e31828cc357. cigarette smoking and APOE genotypes. J. Neurol. Sci. 291 (1–2), 22–29. http://dx.doi.
Lehman, E.J., Hein, M.J., Baron, S.L., Gersic, C.M., 2012. Neurodegenerative causes of death org/10.1016/j.jns.2010.01.011.
among retired National Football League players. Neurology 79 (19), 1970–1974. Schofield, P., Logroscino, G., Andrews, H., Albert, S., Stern, Y., 1997. An association
http://dx.doi.org/10.1212/WNL.0b013e31826daf50. between head circumference and Alzheimer's disease in a population-based study
Logan, S.M., Bell, G.W., Leonard, J.C., 2001. Acute subdural hematoma in a high school of aging and dementia. Neurology 49 (1), 30–37.
football player after 2 unreported episodes of head trauma: a case report. J. Athl. Sephton, C.F., Cenik, C., Kucukural, A., Dammer, E.B., Cenik, B., Han, Y.-H., . . ., Peng, J., 2010.
Train. 36 (4), 433. Identification of neuronal RNA targets of TDP-43-containing ribonucleoprotein com-
Mayeux, R., Ottman, R., Maestre, G., Ngai, C., Tang, M.X., Ginsberg, H., . . ., Shelanski, M., plexes. J. Biol. Chem. 286 (2), 1204–1215.
1995. Synergistic effects of traumatic head injury and apolipoprotein-epsilon 4 in Sephton, C.F., Cenik, B., Cenik, B.K., Herz, J., Yu, G., 2012. TDP-43 in Central Nervous System
patients with Alzheimer's disease. Neurology 45 (3 Pt 1), 555–557. Development and Function: Clues to TDP-43-associated Neurodegeneration.
McAllister, T., Flashman, L., Maerlender, A., Greenwald, R., Beckwith, J., Tosteson, T., . . ., Shively, S., Scher, A.I., Perl, D.P., Diaz-Arrastia, R., 2012. Dementia resulting from traumatic
Duhaime, A.-C., 2012. Cognitive effects of one season of head impacts in a cohort of brain injury: what is the pathology? Arch. Neurol. 69 (10), 1245–1251.
collegiate contact sport athletes. Neurology 78 (22), 1777–1784. Sonnen, J.A., Larson, E.B., Crane, P.K., Haneuse, S., Li, G., Schellenberg, G.D., . . ., Montine,
McKee, A.C., Robinson, M.E., 2014. Military-related traumatic brain injury and neurode- T.J., 2007. Pathological correlates of dementia in a longitudinal, population‐based
generation. Alzheimers Dement. 10 (3), S242–S253. sample of aging. Ann. Neurol. 62 (4), 406–413.
McKee, A., Kosik, K., Kowall, N., 1991. Neuritic pathology and dementia in Alzheimer's Stefansson, H., Helgason, A., Thorleifsson, G., Steinthorsdottir, V., Masson, G., Barnard, J., . .
disease. Ann. Neurol. 30 (2), 156–165. ., Gudnadottir, V.G., 2005. A common inversion under selection in Europeans. Nat.
McKee, A.C., Cantu, R.C., Nowinski, C.J., Hedley-Whyte, E.T., Gavett, B.E., Budson, A.E., . . ., Genet. 37 (2), 129–137.
Stern, R.A., 2009. Chronic traumatic encephalopathy in athletes: progressive Stern, R.A., Daneshvar, D.H., Baugh, C.M., Seichepine, D.R., Montenigro, P.H., Riley, D.O., . . .,
tauopathy after repetitive head injury. J. Neuropathol. Exp. Neurol. 68 (7), 709–735. McHale, L., 2013. Clinical presentation of chronic traumatic encephalopathy. Neurol-
http://dx.doi.org/10.1097/NEN.0b013e3181a9d503. ogy 81 (13), 1122–1129.
McKee, A.C., Gavett, B.E., Stern, R.A., Nowinski, C.J., Cantu, R.C., Kowall, N.W., . . ., Budson, Strain, J., Didehbani, N., Cullum, C.M., Mansinghani, S., Conover, H., Kraut, M.A., . . .,
A.E., 2010. TDP-43 proteinopathy and motor neuron disease in chronic traumatic en- Womack, K.B., 2013. Depressive symptoms and white matter dysfunction in retired
cephalopathy. J. Neuropathol. Exp. Neurol. 69 (9), 918–929. http://dx.doi.org/10. NFL players with concussion history. Neurology 81 (1), 25–32.
1097/NEN.0b013e3181ee7d85. Strickland, D., Smith, S.A., Dolliff, G., Goldman, L., Roelofs, R.I., 1996. Physical activity, trau-
McKee, A.C., Stern, R.A., Nowinski, C.J., Stein, T.D., Alvarez, V.E., Daneshvar, D.H., . . ., Cantu, ma, and ALS: a case–control study. Acta Neurol. Scand. 94 (1), 45–50.
R.C., 2013. The spectrum of disease in chronic traumatic encephalopathy. Brain 136 Strong, M.J., Yang, W., 2011. The frontotemporal syndromes of ALS. Clinicopathological
(Pt 1), 43–64. http://dx.doi.org/10.1093/brain/aws307. correlates. J. Mol. Neurosci. 45 (3), 648–655.
McKee, A.C., Daneshvar, D.H., Alvarez, V.E., Stein, T.D., 2014. The neuropathology of sport. Sullivan, S., Friess, S.H., Ralston, J., Smith, C., Propert, K.J., Rapp, P.E., Margulies, S.S., 2013.
Acta Neuropathol. 127 (1), 29–51. Behavioral deficits and axonal injury persistence after rotational head injury are di-
McQuillen, J.B., McQuillen, E.N., Morrow, P., 1988. Trauma, sport, and malignant cerebral rection dependent. J. Neurotrauma 30 (7), 538–545.
edema. Am. J. Forensic Med. Pathol. 9 (1), 12–15. Talavage, T.M., Nauman, E.A., Breedlove, E.L., Yoruk, U., Dye, A.E., Morigaki, K.E., . . .,
Meaney, D.F., Smith, D.H., Shreiber, D.I., Bain, A.C., Miller, R.T., Ross, D.T., Gennarelli, T.A., Leverenz, L.J., 2014. Functionally-detected cognitive impairment in high school foot-
1995. Biomechanical analysis of experimental diffuse axonal injury. J. Neurotrauma ball players without clinically-diagnosed concussion. J. Neurotrauma 31 (4),
12 (4), 689–694. 327–338.
Miele, V.J., Carson, L., Carr, A., Bailes, J.E., 2004. Acute on chronic subdural hematoma in a The CDC, NIH, DoD, VA Leadership Panel, 2013. Report to Congress on Traumatic Brain In-
female boxer: a case report. Med. Sci. Sports Exerc. 36 (11), 1852–1855. jury in the United States: Understanding the Public Health Problem among Current
Moisse, K., Mepham, J., Volkening, K., Welch, I., Hill, T., Strong, M.J., 2009a. Cytosolic TDP- and Former Military Personnel. Centers for Disease Control and Prevention (CDC),
43 expression following axotomy is associated with caspase 3 activation in NFL−/− the National Institutes of Health (NIH), the Department of Defense (DoD), and the
mice: support for a role for TDP-43 in the physiological response to neuronal injury. Department of Veterans Affairs (VA).
Brain Res. 1296, 176–186. Tremblay, S., Henry, L.C., Bedetti, C., Larson-Dupuis, C., Gagnon, J.-F., Evans, A.C., . . ., De
Moisse, K., Volkening, K., Leystra-Lantz, C., Welch, I., Hill, T., Strong, M.J., 2009b. Divergent Beaumont, L., 2014. Diffuse white matter tract abnormalities in clinically normal age-
patterns of cytosolic TDP-43 and neuronal progranulin expression following ing retired athletes with a history of sports-related concussions. Brain 236 (awu).
axotomy: implications for TDP-43 in the physiological response to neuronal injury. Uryu, K., Giasson, B.I., Longhi, L., Martinez, D., Murray, I., Conte, V., . . ., Trojanowski, J.Q.,
Brain Res. 1249, 202–211. 2003. Age-dependent synuclein pathology following traumatic brain injury in mice.
Montenigro, P.H., Baugh, C.M., Daneshvar, D.H., Mez, J., Budson, A.E., Au, R., Stern, R.A., Exp. Neurol. 184 (1), 214–224.
2014. Clinical subtypes of chronic traumatic encephalopathy: literature review and Uryu, K., Richter-Landsberg, C., Welch, W., Sun, E., Goldbaum, O., Norris, E.H., . . .,
proposed research diagnostic criteria for traumatic encephalopathy syndrome. Trojanowski, J.Q., 2006. Convergence of heat shock protein 90 with ubiquitin in
Alzheimers Res. Ther. 6 (5–8), 1–17. filamentous alpha-synuclein inclusions of alpha-synucleinopathies. Am. J. Pathol.
Morales, R., Green, K.M., Soto, C., 2009. Cross currents in protein misfolding disorders: in- 168 (3), 947–961.
teractions and therapy. CNS Neurol. Disord. Drug Targets 8 (5), 363. Uryu, K., Chen, X.H., Martinez, D., Browne, K.D., Johnson, V.E., Graham, D.I., . . ., Smith, D.H.,
Mori, T., Katayama, Y., Kawamata, T., 2006. Acute hemispheric swelling associated with 2007. Multiple proteins implicated in neurodegenerative diseases accumulate in
thin subdural hematomas: pathophysiology of repetitive head injury in sports. axons after brain trauma in humans. Exp. Neurol. 208 (2), 185–192. http://dx.doi.
Brain Edema XIII. Springer, pp. 40–43. org/10.1016/j.expneurol.2007.06.018.
Mott, F.W., 1916. The effects of high explosives upon the central nervous system. Lancet 1 Vandrovcova, J., Anaya, F., Kay, V., Lees, A., Hardy, J., de Silva, R., 2010. Disentangling the
(12), 331–338. role of the tau gene locus in sporadic tauopathies. Curr. Alzheimer Res. 7 (8),
Mott, F., 1919. War Neuroses and Shell Shock. 726–734.
Nakagawa, A., Manley, G.T., Gean, A.D., Ohtani, K., Armonda, R., Tsukamoto, A., . . ., Waite, L.M., Broe, G.A., Creasey, H., Grayson, D.A., Cullen, J.S., O'Toole, B., . . ., Dobson, M.,
Tominaga, T., 2011. Mechanisms of primary blast-induced traumatic brain injury: 1997. Neurodegenerative and other chronic disorders among people aged 75 years
insights from shock-wave research. J. Neurotrauma 28 (6), 1101–1119. and over in the community. Med. J. Aust. 167 (8), 429–432.
Nordström, P., Michaëlsson, K., Gustafson, Y., Nordström, A., 2014. Traumatic brain injury Yang, Z., Lin, F., Robertson, C.S., Wang, K.K., 2014. Dual vulnerability of TDP-43 to calpain
and young onset dementia: a nationwide cohort study. Ann. Neurol. 75 (3), 374–381. and caspase-3 proteolysis after neurotoxic conditions and traumatic brain injury.
Omalu, B., Hammers, J.L., Bailes, J., Hamilton, R.L., Kamboh, M.I., Webster, G., Fitzsimmons, J. Cereb. Blood Flow Metab. 34 (9), 1444–1452.
R.P., 2011. Chronic traumatic encephalopathy in an Iraqi war veteran with posttrau- Zeng, S., Jiang, J.-X., Xu, M.-H., Xu, L.-S., Shen, G.-J., Zhang, A.-Q., Wang, X.-H., 2014. Prog-
matic stress disorder who committed suicide. Neurosurg. Focus. 31 (5), E3. nostic value of apolipoprotein E epsilon4 allele in patients with traumatic brain inju-
Oppenheimer, D., 1968. Microscopic lesions in the brain following head injury. J. Neurol. ry: a meta-analysis and meta-regression. Gen. Test. Mol. Biomarkers 18 (3), 202–210.
Neurosurg. Psychiatry 31 (4), 299. Zilka, N., Filipcik, P., Koson, P., Fialova, L., Skrabana, R., Zilkova, M., . . ., Novak, M., 2006.
Pearce, A.J., Hoy, K., Rogers, M.A., Corp, D.T., Davies, C.B., Maller, J.J., Fitzgerald, P.B., 2014. Truncated tau from sporadic Alzheimer's disease suffices to drive neurofibrillary de-
Acute motor, neurocognitive and neurophysiological change following concussion generation in vivo. FEBS Lett. 580 (15), 3582–3588.
injury in Australian amateur football. A prospective multimodal investigation. J. Sci.
Med. Sport (In press).
Plassman, B.L., Havlik, R., Steffens, D., Helms, M., Newman, T., Drosdick, D., . . ., Burke, J.,
2000. Documented head injury in early adulthood and risk of Alzheimer's disease
and other dementias. Neurology 55 (8), 1158–1166.

You might also like