You are on page 1of 19

Handbook of Clinical Neurology, Vol.

167 (3rd series)


Geriatric Neurology
S.T. DeKosky and S. Asthana, Editors
https://doi.org/10.1016/B978-0-12-804766-8.00018-2
Copyright © 2019 Elsevier B.V. All rights reserved

Chapter 18

The long-term consequences of repetitive head impacts:


Chronic traumatic encephalopathy

MICHAEL L. ALOSCO1 AND ROBERT A. STERN1,2*


1
Boston University Alzheimer’s Disease and CTE Centers, Department of Neurology, Boston University School of Medicine,
Boston, MA, United States
2
Departments of Neurosurgery, and Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States

Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease associated with exposure to
repetitive head impacts (RHI). Although described in boxers for almost a century, scientific and public
interest in CTE grew tremendously following a report of postmortem evidence of CTE in the first former
professional American football player in 2005. Neuropathologic diagnostic criteria for CTE have been
defined, with abnormal perivascular deposition of hyperphosphorylated tau at the sulcal depths as the
pathognomonic feature. CTE can currently only be diagnosed postmortem, but clinical research criteria
for the in vivo diagnosis of CTE have been proposed. The clinical phenotype of CTE is still ill-defined
and there are currently no validated biomarkers to support an in-life diagnosis of “Probable CTE.” Many
knowledge gaps remain regarding the neuropathologic and clinical make-up of CTE. An increased under-
standing of CTE is critical given the millions that could potentially be impacted by this disease. This chap-
ter describes the state of the literature on CTE. The historical origins of CTE are first presented, followed by
a comprehensive description of the neuropathologic and clinical features. The chapter concludes with
discussion on future research directions, emphasizing the importance of diagnosing CTE during life to
facilitate development of preventative and intervention strategies.

Chronic traumatic encephalopathy (CTE) is a distinct (Stern et al., 2013; Montenigro et al., 2014). Despite
neurodegenerative disease associated with a history of these advancements, research on CTE remains in its
exposure to repetitive head impacts (RHI), such as those infancy, and many knowledge gaps need to be addressed
incurred by contact sport athletes and military veterans given the millions of individuals who could potentially
(McKee et al., 2013, 2016; Bieniek et al., 2015; Ling be impacted by this devastating, but possibly prevent-
et al., 2017; Mez et al., 2017; Tagge et al., 2018; able, disease. The purpose of this chapter is to provide
Falcon et al., 2019; Stern et al., 2019). Chronic traumatic a comprehensive review on the current state of the liter-
encephalopathy may be a major public health concern; ature on CTE in order to convey what is currently known
however, the epidemiology of CTE remains unknown and not known about CTE. We present the historical
because this disease can currently only be diagnosed origins of CTE briefly, followed by a detailed description
postmortem using recently defined neuropathologic of its neuropathologic features. The role of RHI in the
diagnostic criteria (McKee et al., 2016). Chronic trau- development of CTE is discussed, and a review of the
matic encephalopathy has indeed become well-defined clinical presentation and features of CTE is provided.
neuropathologically over the past decade, and clinical The chapter concludes with a discussion on directions
research diagnostic criteria have also been proposed for future research.

*Correspondence to: Robert A. Stern, Boston University Alzheimer’s Disease and CTE Centers, Department of Neurology,
Boston University School of Medicine, 72 E. Concord St, Suite B7800, Boston, MA 02118, United States. Tel: +1-617-358-5375,
Fax: +1-617-358-6544, E-mail: bobstern@bu.edu
338 M.L. ALOSCO AND R.A. STERN
CHRONIC TRAUMATIC “Chronic Traumatic Encephalopathy in a National Foot-
ENCEPHALOPATHY: HISTORICAL ball League Player” (Omalu et al., 2005). Since that time,
ORIGINS CTE has increasingly become a household term due to
neuropathologic evidence of this disease in many ensu-
The historical origins of CTE have been described in
ing well-known American professional football players
detail by Montenigro et al. (2015). CTE dates back to
(e.g., Junior Seau, John Mackey, Dave Duerson), as well
1928, when Harrison Martland introduced the term
as an array of other modern day contact sport athletes
“Punch Drunk” to describe his observation of a clinical
(e.g., ice hockey, soccer, boxing, rugby), soldiers
syndrome in prizefighters. The prizefighters exhibited
deployed to Iraq and Afghanistan, and civilians with
early onset behavioral disturbances, characterized by
recurrent head trauma (McKee et al., 2013; Ling et al.,
Martland as “goofy,” “slug nutty,” and “cuckoo,” fol-
2017; Lee et al., 2019).
lowed by later onset “mental deterioration” (Martland,
1928). Following Martland’s paper, numerous different
nosologic terms were used throughout the years to refer
NEUROPATHOLOGIC FEATURES OF
to a constellation of behavior, mood, cognitive, and
CHRONIC TRAUMATIC
motor symptoms found in boxers, including the well-
ENCEPHALOPATHY
known term “dementia pugilistica,” which was coined
in a 1937 publication that simultaneously provided Corsellis et al. (1973) and Omalu et al. (2005) were
neuropathologic support for the disease (Millspaugh, among the first to describe the neuropathologic features
1937). The term “chronic traumatic encephalopathy” of CTE. Through the case reports from Omalu (Omalu
was designated in 1940 in the description of a profes- et al., 2010a,b, 2011), and the extensive, ongoing studies
sional boxer with chronic behavior/mood and cognitive by McKee and colleagues at the Boston University (BU)
disturbances (Bowman and Blau, 1940). In 1949, British Alzheimer’s Disease and CTE Centers and the Veterans
neurologist Macdonald Critchley again used the term Administration-BU–Concussion Legacy Foundation
CTE in the context of symptomatic boxers. Neuropatho- (VA-BU-CLF) Brain Bank (McKee et al., 2013), the neu-
logic research in the 1950s and 1970s began to provide ropathologic descriptions of CTE have been refined.
support for a relationship between boxing and damage Through an ongoing study funded by the National Insti-
to regions of the brain, including the septum pellucidum, tute of Neurological Disorders and Stroke (NINDS),
periventricular gray matter, frontal and temporal lobes, entitled, “Understanding Neurologic Injury in Traumatic
and substantia nigra, as well as cerebellar scarring and Encephalopathy (UNITE)” (PI: Ann McKee) (Mez et al.,
neuronal loss (Brandenburg and Hallervorden, 1954; 2015), neuropathologic diagnostic criteria for CTE have
Corsellis et al., 1973). Corsellis et al. (1973) observed been examined during a recent NINDS and National
widespread p-τ neurofibrillary tangles (NFT) in the Institute of Biomedical Imaging (NIBIB) sponsored con-
cerebrum and brain stem in their case series study of ference. A panel of seven neuropathologists with exper-
15 retired boxers. Notably, in the remote research show- tise in neurodegenerative diseases, in general, and
ing the long-term clinical consequences of boxing, tauopathies, in particular, convened to evaluate McKee
there is also mention of American football as a potential et al.’ (2013) neuropathologic criteria for CTE. Each
source of chronic symptom sequelae (Osnato and neuropathologist evaluated 25 selected cases of various
Giliberti, 1927; Carroll, 1936; Bowman and Blau, tauopathies, with CTE cases being representative of
1940; Martland, 1943). the disease and being at least of moderate disease sever-
It was not until 2005 that CTE became a societal ity. There was good agreement for overall neuropatho-
concern after neuropathologic changes consistent with logic diagnosis (k ¼ 0.67) and even better for CTE
CTE were found in a former National Football League diagnosis (k ¼ 0.78). The neuropathologic criteria agreed
(NFL) player. Mike Webster was a former lineman for upon for diagnosing CTE are presented in Table 18.1.
the Pittsburgh Steelers and Kansas City Chiefs who The conference led to a consensus agreement that CTE
experienced depression and memory problems after retir- is a “unique disease” caused, in part, by RHI, with the
ing from football and was demented prior to his cardiac- pathognomonic lesion being the deposition of p-τ neuro-
related death in 2002 at the age of 50 (Fainaru-Wada and fibrillary, astrocytic, and neuropil tangles around small
Fainaru, 2013). Neuropathologist Bennet Omalu and his blood vessels in an irregular pattern at the depths of
colleagues examined Webster’s brain at postmortem and the cerebral sulci (see Fig. 18.1)—a pattern unique from
discovered it to be macroscopically normal, but micro- any other tauopathy, including Alzheimer’s disease (AD)
scopically abnormal due to evidence of hyperpho- and frontotemporal dementia (FTD). Recent work also
sphorylated tau (p-τ). Omalu published this case in the shows that the specific tau species of CTE is unique
journal Neurosurgery in 2005, in an article entitled, (Falcon et al., 2019).
Table 18.1
Neuropathologic criteria for diagnosing chronic traumatic encephalopathy (McKee et al., 2016)

Required for diagnosis of CTE


The pathognomonic lesions consists of p-τ aggregates in neurons, astrocytes, and cell processes around small vessels in an irregular
pattern at the depths of the cortical sulci
Supportive neuropathologic features of CTE
P-τ-related pathologies

1. Abnormal p-τ immunoreactive pretangles and NFTs preferentially affecting superficial layers (layers II–III), in contrast to layers
III and V as in AD
2. In the hippocampus, pretangles, NFTs, or extracellular tangles preferentially affecting CA2 and pretangles and prominent
proximal dendritic swellings in CA4. These regional p-τ pathologies differ from the preferential involvement of CA1 and
subiculum found in AD
3. Abnormal p-τ immunoreactive neuronal and astrocytic aggregates in subcortical nuclei, including the mammillary bodies and
other hypothalamic nuclei, amygdala, nucleus accumbens, thalamus, midbrain tegmentum, and isodendritic core (nucleus basalis
of Meynert, raphe nuclei, substantia nigra, and locus coeruleus)
4. P-τ immunoreactive thorny astrocytes at the glial limitans most commonly found in the subpial and periventricular regions
5. P-τ immunoreactive large grain-like and dot-like structures (in addition to some threadlike neurites)
Non-p-τ related pathologies

1. Macroscopic features: disproportionate dilatation of the third ventricle, septal abnormalities, mammillary body atrophy, and
contusions or other signs of previous traumatic injury
2. TDP-43 immunoreactive neuronal cytoplasmic inclusions and dot-like structures in the hippocampus, anteromedial temporal
cortex, and amygdala
Age-related p-τ astrogliopathy that may be present; nondiagnostic and nonsupportive

1. Patches of thorn-shaped astrocytes in subcortical white matter


2. Subependymal, periventricular, and perivascular thorn-shaped astrocytes in the mediobasal regions
3. Thorn-shaped astrocytes in amygdala or hippocampus

Table borrowed from McKee, A.C., Cairns, N.J., Dickson, D.W., et al., 2016. The first NINDS/NIBIB consensus meeting to define neuropatho-
logical criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol 131: 75–86 under the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/legalcode).
No text changes were made, but the title was changed and the format of the table was revised.

Fig. 18.1. Pathognomonic lesion of chronic traumatic encephalopathy. No changes were made to the illustration; however, minor
changes were made for brevity to the below legend. (A) Perivascular p-τ lesion at the sulcal depths. (B–F) Multiple perivascular
foci in the cortex in CTE. (G) The p-τ aggregates in CTE are rounded structures in the neuropil that are most dense in the areas
surrounding the vessel. (H) The rounded p-τ immunoreactive cell processes are more densely distributed than those found in argyr-
ophilic grain disease. Figure borrowed from McKee, A.C., Cairns, N.J., Dickson, D.W., et al., 2016. The first NINDS/NIBIB con-
sensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol 131:
75–86 under the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/legalcode).
340 M.L. ALOSCO AND R.A. STERN
McKee et al. (2013) proposed a 4-stage classification Axonal pathology
system to grade the pathologic severity and progression
There is axonal injury, axonal degeneration, myelinated
of p-τ in CTE, with stage I being the least severe and
fiber loss, and white matter atrophy in every stage of
stage IV being the most severe. As described in the fol-
CTE. Axonal pathology corresponds to the progression
lowing text, the stages are accompanied by progressively
of neural degeneration. In stages I and II, there are scat-
worsening macro- and microscopic brain changes, axo-
tered and distorted axonal varicosities in the frontal and
nal pathology, chronic neuroinflammation, and 43 kDa
temporal cortices, as well as in the subcortical white mat-
TAR DNA-binding protein (TDP-43) abnormalities.
ter and deep white matter tracts of the diencephalon. As
pathologic severity advances (stages III and IV), there is
MACROSCOPIC PATHOLOGY IN CTE striking axonal loss and distorted axonal profiles in the
Gross pathology is unremarkable in stage I CTE. In stage cortex and subcortical white matter pathways, as well
II, various macroscopic pathologies are present, includ- as white matter macrophages and p-τ abnormalities.
ing mild enlargement of the lateral ventricles, mild
enlargement of the third ventricle, sharp concavity of TDP-43
the third ventricle, cavum septum pellucidum, and pallor
of the locus coeruleus and/or substantia nigra. In stage III TDP-43 is an RNA-protein (Sephton et al., 2011, 2012)
CTE, there is mild cerebral atrophy and dilation of the that binds to tau (Sato et al., 2009) and influences tau
lateral and third ventricles. Approximately 40% of stage isoform expression (Morales et al., 2009). Stage I CTE
III cases exhibit septal abnormalities (e.g., cavum sep- cases often have TDP-43 neuritic thread and dot-like
tum, septal perforations), nearly 60% have moderate inclusions in the subcortical white matter and fornix.
depigmentation of the locus coeruleus, and 40% of the TDP-43 inclusions are present in stage II and III CTE
brains show mild pallor of the substantia nigra. Thus, cases and are located in the subcortical white matter,
neurotransmitter dysfunction or loss is clear and is added brainstem, or MTL (stage II), as well as in the cerebral
to the other atrophic and connectivity losses. In addition, cortex (stage III). By stage IV, TDP-43 immunopositivity
atrophy of the mammillary bodies and thalamus, thinning can be widespread throughout the cerebrum, MTL,
of the hypothalamic floor and corpus callosum, and con- brainstem, diencephalon, basal ganglia, and occasion-
vex contour of the medial thalamus may be present. In ally, the spinal cord.
stage IV CTE, there is marked shrinkage of the cortex,
white matter, medial temporal lobe (MTL), thalamus, b-Amyloid
hypothalamus, and mammillary bodies. A majority of
The presence of b-amyloid neuritic plaques is a hallmark
stage IV CTE brains exhibit pallor of the locus coeruleus
pathology of AD, and is part of the diagnostic criteria for
and substantia nigra, in addition to enlargement of the
AD as per both the National Institutes of Aging (NIA)-
lateral ventricles, enlargement or sharp concavity of
Reagan Institute and the NIA-Alzheimer’s Association
the third ventricle, and septal abnormalities.
guidelines (Newell et al., 1999; Montine et al., 2012).
Unlike AD, b-amyloid peptide deposits are not a consis-
MICROSCOPIC PATHOLOGY IN CTE tent pathologic feature of CTE, and if present, they are dif-
P-τ fuse and dense core plaques (McKee et al., 2013). In a
2015 study, b-amyloid was found in 52% of former con-
Stage I CTE involves focal perivascular epicenters of tact sport athletes and military veterans with neuropatho-
p-τ NFT and astrocytic tangles at the sulcal depths, logically confirmed CTE and was associated with older
particularly in the superior, dorsolateral, and inferior age and the apolipoprotein E (APOE) E4 allele (Stein
frontal cortices. In stage II, p-τ NFT become dispersed et al., 2015b); carriers of this allele are known to have
throughout the cortex and extend into the superficial greater accumulation of b-amyloid than noncarriers.
layers of the cortex adjacent to the perivascular epicen- b-amyloid deposition may thus occur at an accelerated
ters formed in stage I CTE. There are moderate NFT rate in the presence of CTE pathology.
densities in the nucleus basalis of Meynert and locus
coeruleus. P-τ NFT spread further throughout the
COMORBID NEUROPATHOLOGY
cerebral cortex in stage III, and appear in the MTL
(e.g., hippocampus, entorhinal cortex, amygdala), olfac- CTE is frequently comorbid with other neurodegenera-
tory bulb, mammillary bodies, diencephalon, brainstem, tive diseases, including Lewy body disease (LBD),
and spinal cord. In stage IV, p-τ is widely dispersed AD, motor neuron disease (MND), and frontotemporal
throughout the cortex, MTL, diencephalon, basal lobar degeneration (FTLD) (McKee et al., 2013; Mez
ganglia, brainstem, and spinal cord. et al., 2017). The presence of one disease may increase
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 341
risk for another, possibly due to the interaction between and describe their associated prevalence and symptom
various proteins (e.g., tau, a-synuclein) (Jellinger, 2012; phenotype. Evidence supporting RHI as a necessary
Stein et al., 2014). The comorbidity between CTE and risk factor for CTE is then presented, and possible
AD is noteworthy given the extant research associating mechanisms for this relationship are discussed.
traumatic brain injury (TBI), in particular moderate
and severe TBI, with increased risk for AD (Fleminger REPETITIVE HEAD IMPACTS:
et al., 2003). Autopsy and in vivo positron emission TERMINOLOGY AND PREVALENCE
tomography (PET) studies have linked moderate to
Concussion
severe TBI with increased b-amyloid burden (Johnson
et al., 2012; Scott et al., 2016). It has been theorized that Concussion, a mild TBI subtype, is a complex patho-
CTE is and has been clinically misdiagnosed as AD in the physiologic process affecting the brain secondary to
setting of TBI or recurrent head trauma (Lehman et al., biomechanical forces induced from direct or indirect
2012) given that most studies examining the association blows to the head (McCrory et al., 2013). An estimated
between TBI and AD risk have been without neuropath- 1.6–3.8 million concussions occur from sports and recre-
ologic confirmation of AD as the etiology. Notably, rel- ational activities each year in the United States (Langlois
ative to a non-TBI AD group, AD subjects with a history et al., 2006). The true prevalence of concussion is likely
of TBI have been shown to exhibit clinical features con- much greater, given that many or most are not reported.
sistent with CTE and have lower amyloid angiopathy at Sport-related concussion rates are highest in American
autopsy (Sayed et al., 2013). Recent studies that have football, but are prevalent in other contact sports as well
included autopsy and/or in vivo biomarker evidence of (e.g., ice hockey, lacrosse, soccer) (Daneshvar et al.,
AD have also failed to find an association between 2011). Additionally, mild TBI is common in military per-
TBI and AD pathology (Crane et al., 2016; Weiner sonnel. In 2014, the Department of Defense estimated the
et al., 2017; Sugarman et al., 2019). number of new worldwide medical diagnoses of TBI
A subset of CTE cases exhibit motor neuron symp- in veterans to be approximately 25,044, with just over
toms (e.g., muscle weakness, atrophy, spasticity, and fas- 20,000 being mild (Defense and Veterans Brain Injury
ciculations) that mimic Amyotrophic Lateral Sclerosis Center, 2015). Typical classification criteria for concussion
(ALS) (McKee et al., 2010, 2013). CTE subjects with and mild TBI include post-injury confusion, disorientation,
comorbid MND die at a younger age (CTE stages II–III), or loss of consciousness (LOC)  30 min; a Glasgow Coma
and MND in CTE may be a consequence of TDP-43 pro- Scale score of 13–15; and/or posttraumatic amnesia <24 h
teinopathy in the brain and spinal cord (McKee et al., (Kay et al., 1993). Absence of structural brain alterations
2010, 2013). To a lesser extent, CTE also cooccurs with on standard CT or magnetic resonance imaging (MRI) is
FTLD. CTE has also been characterized as an FTLD characteristic of concussion and mild TBI.
subtype due to the nature (e.g., tau and TDP-43 proteins) Concussion symptoms typically fall into three
and topographic deposition of pathology (e.g., fronto- domains: neurologic, behavioral, and psychiatric
temporal) in CTE (McKee et al., 2010). Finally, recent (Ponsford et al., 2000; McCrory et al., 2013). Neurologic
work suggests dot-like lesions as a possible significant symptoms can include headache, dizziness, sensitivity to
feature of CTE, which may have implications for argyr- light and noise, tinnitus, and vision disturbances. Cog-
ophilic grain disease and Parkinson’s disease dementia nitive problems can appear immediately after the con-
(Armstrong et al., 2017). cussion, such as difficulties in attention, psychomotor
speed, and memory. Behavioral and psychiatric symptoms
commonly involve depression, insomnia, fatigue, and irri-
THE ROLE OF REPETITIVE HEAD
tability. A majority of symptomatic concussions resolve
IMPACTS IN CHRONIC TRAUMATIC
in days to weeks (Belanger and Vanderploeg, 2005;
ENCEPHALOPATHY
McCrory et al., 2013), but in some cases, symptoms
Repetitive head impacts refer to the cumulative lifetime may persist for up to 3 months following the injury
exposure of an individual to recurrent concussive (or (Ponsford et al., 2000; Belanger et al., 2005; Iverson,
mild TBI) and subconcussive injuries (Montenigro 2005). In a small subset of individuals, psychologic, cog-
et al., 2016). RHI can also include recurrent moderate nitive, and somatic symptoms following concussion can
and severe TBIs, but the majority of all head trauma is last 1 year or longer (Katz and DeLuca, 1992; Dikmen
mild in nature. CTE has been studied and characterized et al., 2001; Bigler, 2008; W€aljas et al., 2015; Losoi
largely in the context of exposure to sport-related concus- et al., 2016). Recurrent concussive injuries may increase
sive and subconcussive head impacts, and to a lesser risk for more severe future concussive injuries, exacerbate
extent, head trauma in military veterans. In the following symptom presentation, and delay recovery (Collins et al.,
text, we first define concussion and subconcussive injury 2002; Guskiewicz et al., 2003; Iverson et al., 2004).
342 M.L. ALOSCO AND R.A. STERN
Subconcussive injury is associated with worse CTE neuropathologic severity
(McKee et al., 2013; Alosco et al., 2018a,b).
A subconcussive injury involves a direct or indirect blow
Bieniek et al. (2015) reviewed medical records of
to the head that results in the transfer of mechanical energy
>1700 brain donation cases from the Mayo Clinic brain
to the brain at a force sufficient to injure axonal integrity,
bank to ascertain history of amateur contact sport partic-
but does not result in overt clinical symptoms of concus-
ipation. The authors evaluated the brains of 66 donors
sion (Dashnaw et al., 2012; Bailes et al., 2013; Belanger
identified to have had exposure to contact sports in high
et al., 2015; Erlanger, 2015). The threshold intensity level
school or college (34 played American football) for evi-
of impact that causes neuronal damage but does not result
dence of CTE pathology, and applied the staging system
in clinical symptoms remains unknown. Subconcussive
proposed by McKee et al. (2013). The authors found that
events are common in the setting of contact sports, and
21 of the 66 former contact sport athletes exhibited neu-
helmet-based accelerometer studies estimate that amateur
ropathologic changes consistent with the disease (e.g.,
high school football players average 600 subconcussive
perivascular cortical p-τ). Remarkably, CTE pathology
impacts with a seasonal linear acceleration of 16,746.1g
was not present in 198 age- and disease-matched men
each season, an estimate that increases to over 1000 at
and women without exposure to contact sports, including
the collegiate level (Crisco et al., 2010; Broglio et al.,
33 with a single TBI sustained from nonathletic causes
2011; Gysland et al., 2012). Although subconcussive head
(e.g., falls, motor vehicle accidents). Ling et al. (2015)
impacts do not result in immediate symptoms, repetitive
also examined 268 neurodegenerative and control cases
subconcussive blows registering greater than 14.4g on
from the Queen Square Brain Bank for Neurological
helmet accelerometer sensors have been linked with
Disorders for evidence of CTE pathology. CTE neuropa-
pre- to postseason cognitive decline, functional brain
thology was found in 11.9% of neurodegenerative disor-
alterations, and white matter changes in high school foot-
ders and 12.8% of elderly controls. Of these cases, 93.8%
ball players (Davenport et al., 2014; Talavage et al., 2014;
had a history of TBIs, 34% had a history of participation
Abbas et al., 2015). Additional studies of hockey (Koerte
in contact sports, and 18.8% were military veterans.
et al., 2012a) and soccer players (Koerte et al., 2012b,
Research has also found evidence of CTE (and other
2015a; Lipton et al., 2013) have demonstrated altered
neurodegenerative disease pathologies) in former soccer
white matter integrity and brain biochemistry associated
plays without a known history of concussion, suggesting
with subconcussive exposure.
a possible link between repetitive subconcussive injuries
from routine heading and long-term neurologic disorders
(Ling et al., 2017).
REPETITIVE HEAD IMPACTS
Recurrent concussion is a component of RHI, but it REPETITIVE HEAD IMPACTS AND CTE:
is the numerous lifetime subconcussive head impacts POTENTIAL MECHANISMS
that appear to play a prominent role in the pathogenesis The mechanisms underlying the association between
of CTE. For example, 16% of neuropathologically RHI and CTE are not currently known and are likely
confirmed cases of CTE have no history of reported multifaceted. The acceleration and deceleration of the
concussions, but did have significant subconcussive head during concussion can result in shearing and
exposure (Stein et al., 2015a). Furthermore, McKee tensile forces on long fibers, such as axons and blood
et al. (2013) concluded that family-reported number of vessels, and cause traumatic axonal injury and an
concussions was unrelated to CTE pathologic stage, array of neurometabolic pathophysiologic changes
yet all 68 cases diagnosed with CTE had a history of sub- (e.g., ionic flux and glutamate release, neuroinflamma-
concussive trauma. Of the 68 individuals diagnosed with tion, altered neurotransmission, microvasculopathy)
CTE 64 were contact sport athletes. (The remaining four (Maxwell et al., 1997; Medana and Esiri, 2003; Giza
were three military veterans, and one individual with a and Hovda, 2014). Areas of the brain that are susceptible
history of self-injurious head banging.) These included to axonal direction and/or tissue-density change (e.g.,
50 football players (34 professional), 5 ice hockey around small blood vessels, gray–white matter interface)
players (enforcers), 7 professional and 1 amateur boxer, are most sensitive to the biomechanical rotational
and 1 professional wrestler. The 18 age- and gender- forces of concussion (Oppenheimer, 1968; Povlishock,
matched controls without history of RHI exposure were 1993; Blumbergs et al., 1994; Gaetz, 2004; Cloots
absent CTE neuropathology. Considering that many pro- et al., 2011; McKee et al., 2014). Experimental studies
fessional athletes begin contact sport participation as in animals demonstrate that axonal injury and vascular
youth, they likely experience thousands of lifetime subcon- disruption following RHI is most severe at focal stress
cussive head impacts. Moreover, longer duration of expo- points such as the perivascular regions and depths of
sure to RHI (as operationalized by years of football play) cortical sulci (Smith et al., 1999; Cloots et al., 2008).
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 343
RHI-related axonal injury may also contribute to the tau only one exposure to blast and was without any previous
protein, a stabilizer of axonal microtubules, becoming or postmilitary service TBI history. Single blast exposure
abnormally misfolded, phosphorylated, and aggregated and CTE pathology has also been observed in mice
and cleaved, leading to neurotoxic tau fragments and (Goldstein et al., 2012). Multiple exposures may be
neuronal death (McKee et al., 2013, 2014, 2015; occurring within a single blast due to cycles of oscilla-
Mouzon et al., 2014). tion in head acceleration–deceleration, known as the
It is unclear how the acute pathophysiologic changes “bobblehead effect” (Goldstein et al., 2012). For a com-
associated with RHI (e.g., axonal injury, neuroinflamma- prehensive review on blast exposure in military veterans
tion, blood–brain barrier breach, disruption of tau) tran- and CTE, see Goldstein and colleagues (2014).
sition to the progressive neural degeneration observed in
CTE. Blaylock and Maroon (2011) theorized that it may
involve chronic immunoexcitotoxicity activity. In a nor-
CLINICAL PRESENTATION OF CHRONIC
mally functioning individual, microglia aid in immune
TRAUMATIC ENCEPHALOPATHY
defense of the central nervous system and serve to
decrease neuroinflammation and repair neuronal func- Clinical presentation
tion. However, activated microglia in the presence of
POSTMORTEM RETROSPECTIVE DATA
brain pathology can be neurodestructive and remain so
for a prolonged period of time. As hypothesized by McKee et al. (2013) provided an initial description of
Blaylock and Maroon (2011), an initial head injury the clinical symptoms associated with CTE that included
activates (or primes) microglia, and the microglia remain a combination of behavior (e.g., aggression, explosivity),
activated for extended periods of time. Subsequent head mood (e.g., depression, suicidal ideations), and cognitive
injuries cause the microglia to become hyperactive, symptoms (e.g., executive dysfunction, episodic mem-
resulting in massive releases of immune cytokines and ory impairment). These findings were consistent with
excitotoxin neurochemicals (i.e., glutamate). The white earlier reports of boxers (Mawdsley and Ferguson,
matter degeneration (gliosis) (Blaylock and Maroon, 1963; Corsellis et al., 1973). Stern et al. (2013)
2011; Smith et al., 2013), and chronic astrocytosis and conducted a comprehensive study to better delineate
microgliosis that may occur during RHI exposure can the clinical nature and course of symptoms in CTE.
also lead to glutamate accumulation (Ojo et al., 2013; Retrospective next-of-kin telephone interviews were
Mannix et al., 2014). Glutamate continues to be released conducted for 36 male contact sport athletes (29 football
as microglial activation and axonal injury persist players, 3 professional ice hockey players, 1 professional
with each subsequent head injury. The result is chronic wrestler, and 3 boxers) who donated their brain to
immunoexcitotoxicity that could precipitate neural the VA-BU-CLF Brain Bank, were neuropathologically
degeneration. Some of these neuroinflammatory and diagnosed with CTE, and were without any other
vascular-based mechanisms were recently supported significant comorbid neuropathology. Next-of-kin
by an experimental study (Tagge et al., 2018). interviews were conducted blinded to the neuropathol-
ogy of the brain donor, and neuropathologists were
blinded to all clinical data. The results revealed two
MILITARY VETERANS AND CHRONIC
distinct clinical presentations: (1) initial onset of behav-
TRAUMATIC ENCEPHALOPATHY
ioral (e.g., explosivity, impulsivity, aggression) or
A majority of neuropathologically diagnosed cases of mood (e.g., depression) changes at a young age (mean
CTE have been former contact sport athletes, but military age ¼ approximately 35 years old) and (2) later-life onset
veterans may also be at risk. McKee et al. (2013) found (mean age ¼ approximately 60 years old) of cognitive
16 of 21 military veterans with CTE were also contact impairment, particularly in episodic memory and execu-
sport athletes (8 former NFL players) and 9 of the vet- tive function. The subjects with initial behavior or mood
erans saw combat (4 in Iraq and Afghanistan, 1 in Gulf symptoms were combined into a single behavior/mood
War, 2 in Vietnam, and 2 in World War II), and 3 were group due to similarities in age of symptom onset, age
exposed to blast. A history of blast exposure in military at death, and neuropathologic stage. There was a sig-
veterans is of particular concern in terms of CTE risk. nificant delay (years to decades) between end of RHI
Postmortem analysis of a sample of five brains of US exposure and symptom onset. All subjects demons-
male military veterans with history of blast exposure trated cognitive impairment at some point, but the cog-
revealed the presence of the pathognomonic CTE lesion nitive subgroup was less likely to have a history of
that was indistinguishable from CTE neuropathology in behavioral/mood symptoms and usually developed
age- and gender-matched contact sport athletes with a dementia (i.e., cognitive impairment significant enough
history of RHI (Goldstein et al., 2012). CTE neuropa- to impact functional independence). The cognitive
thology was present for one military veteran who had subgroup was more advanced in disease severity.
344 M.L. ALOSCO AND R.A. STERN
In the study by Stern et al. (2013), none of the subjects of former NFL players relative to demographically-
had motor features as an initial presenting feature, and matched controls (Hart et al., 2013), and executive
overall, motor symptoms were not common. This is in dysfunction has been reported in a sample of active
contrast to the earlier reports on boxers who described and former college and professional football players
motor features to be a prominent symptom (Mawdsley (Seichepine et al., 2013).
and Ferguson, 1963; Corsellis et al., 1973) and, as previ-
ously described, the established comorbidity of MND
CLINICAL RESEARCH DIAGNOSTIC
and CTE. Although unclear, boxers may be more prone
CRITERIA
to develop parkinsonism due to the angular acceleration
and torsional focal stress placed on the brainstem and cer- Three different author groups (Jordan, 2013; Victoroff,
ebellum, which is different from the transverse and linear 2013; Montenigro et al., 2014) have proposed similar
acceleration and deceleration biomechanical forces in clinical research diagnostic criteria that are compared
football (Viano et al., 2005; Beitz, 2014). Consistent with in detail elsewhere (Baugh et al., 2014). The primary dif-
this notion, McKee et al. (2013) observed that motor ference among the criteria is the central role of motor fea-
symptoms and cerebellar pathology were more prevalent tures for the clinical diagnosis of CTE in the Jordan
in former boxers relative to former football players. (2013) criteria, whereas motor symptoms are supportive
Motor impairment may be an initial presenting symptom features of CTE in the other criteria. Montenigro et al.
in boxers due to earlier degeneration of brain regions like (2014) coined the nosology “Traumatic Encephalopathy
the substantia nigra. In contrast, motor symptoms in foot- Syndrome (TES)” to refer to the clinical syndrome asso-
ball players might not manifest until later in the disease ciated with CTE (as well as other possible etiologies with
course in the context of widespread neural degeneration. brain trauma exposure) and suggested to reserve the term
“CTE” for the postmortem diagnosis. The TES diagnos-
tic criteria are based on a systematic review of the clinical
IN VIVO RESEARCH
features of 83 neuropathologically confirmed CTE cases
Chronic traumatic encephalopathy cannot currently be in the literature. The TES diagnosis requires a history of
diagnosed during life partially due to the lack of vali- multiple impacts to the head, such as multiple mild, mod-
dated biomarkers. However, in vivo research has been erate, and/or severe TBIs, or exposure to concussive and
examining the clinical phenotype of subjects presumably subconcussive events. Exposure sources could include,
at high risk for CTE (e.g., former NFL players). These but are not limited to, contact sports, military service,
studies corroborate behavioral/mood and cognitive domestic violence, and/or head-banging, among others.
impairment as core features of CTE. In an independent Consistent with Stern et al. (2013), Montenigro et al.
study at the University of California, San Francisco, (2014) found three core clinical features to most com-
Gardner et al. (2015) conducted a case series study of monly emerge during the years to decades after RHI
14 symptomatic, former professional male American exposure, with the presence of one or more of the follow-
football players presenting to an academic memory ing three core clinical features necessary for diagnosis:
clinic. Three of the subjects exhibited delayed-onset (1) behavioral symptoms primarily characterized by
behavioral/mood symptoms (e.g., anxiety, depression, aggression and impulsivity; (2) mood dysfunction
irritability, impulsivity), and five had delayed-onset pro- characterized by depression and related symptoms
gressive cognitive symptoms (e.g., memory impairment, (e.g., hopelessness, suicidality); and (3) cognitive diffi-
executive dysfunction), one of whom displayed an initial culties that involve cognitive decline and impaired cog-
motor symptom onset with cognitive symptoms emerg- nitive test performance (i.e., 1.5 SD below the normative
ing 5 years later. There was also a third phenotype that mean) in attention, executive function, and/or episodic
included five football players who exhibited chronic memory. These core features comprise four distinct
postconcussive symptoms (e.g., headache, forgetfulness, diagnostic TES variants: TES behavioral/mood variant
concentration difficulties, depression, apathy) that (TES-BMv), TES cognitive variant (TES-COGv), TES
began during their playing career. Neuropsychologic mixed variant (TES-MIXv), and TES dementia
testing of all subjects revealed verbal and visual memory (TES-D). TES-D requires a progressive course of cogni-
impairment and executive dysfunction to be common tive core features and evidence of functional impairment.
and most prominent in the cognitive group. All of the There must also be a minimum of two of the following
behavior/mood subjects had an elevated Geriatric supportive features: impulsivity, anxiety, apathy, para-
Depression Scale score, whereas only 17% of the cogni- noia, suicidality, headache, motor signs, documented
tive/motor group showed clinically meaningful symp- decline (decline in function, symptoms, or signs based
toms of depression. Cognitive impairment, dementia, on formal measurement), or delayed onset (at least
and depression have also been found in other samples 2 years after RHI exposure). Modifiers include the
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 345
presence of motor features (e.g., dysarthria, dysgraphia, sample of former American professional football players
bradykinesia) and the nature of the clinical course (Gardner et al., 2016), and 12 of the 14 former profes-
(stable, progressive, unknown/inconsistent). sional football players from the Gardner et al. (2015) case
A separate diagnostic classification for “Possible series study had a CSP.
CTE,” “Probable CTE,” and “Unlikely CTE” is included
in the TES criteria and is based on the presence of poten- MICROSTRUCTURAL CHANGES
tial biomarkers, which, when validated, indicate the
Diffusion tensor imaging (DTI) is an MRI technique
degree to which the underlying etiology of the clinical
sensitive to water diffusion within brain tissue. It is com-
presentation of TES is due to the CTE pathophysiologic
monly used to assess microstructural axonal integrity
process. The TES diagnosis can be designated in the
because water movement is directionally dependent on
presence of other neurodegenerative conditions other
the fiber tracts in white matter (Ng et al., 2014). There
than CTE who meet the requisite exposure and symptom
is significant white matter pathology in every stage of
criteria. It is therefore critical that research identify and
CTE, and DTI may be a useful tool to detect CTE-related
validate in vivo biomarkers to facilitate the diagnosis
white matter pathology due to its sensitivity to axonal
of “Probable CTE.”
injury in concussion (Shenton et al., 2012). Many studies
have demonstrated reduced microstructural white matter
POTENTIAL BIOMARKERS OF CTE integrity in active contact sport athletes (Zhang et al.,
The use of biomarkers has become the gold standard for 2003; Chappell et al., 2006; Koerte et al., 2012b;
diagnosing “Probable AD” during life (Jack et al., 2011; Lipton et al., 2013; Bazarian et al., 2014), and only lim-
McKhann et al., 2011), and a similar approach is being ited research has examined RHI exposure and long-term
adopted in CTE. The Montenigro et al. (2014) TES clin- white matter changes. Reduced white matter integrity in
ical research criteria proposed the following biomarkers the frontal lobe predicted severity of depressive symp-
to designate “Probable CTE”: Cavum septum pelluci- toms in a sample of 26 former NFL players (Strain
dum, normal b-amyloid cerebrospinal fluid (CSF) levels, et al., 2013). White matter integrity of the frontal lobe
elevated CSF p-τ/τ ratio, negative amyloid imaging, pos- and its subcortical connections may be of particular inter-
itive tau imaging, cortical thinning, and cortical atrophy. est given the frontal distribution of pathology in CTE and
Although limited, the extant research on the potential the frontal-mediated symptoms observed in this disease.
utility of these biomarkers in CTE, as well as others that There is also support in the animal literature for long-
have been identified since the proposal of TES, are dis- term white matter injury subsequent to repetitive cortical
cussed in the following text. Of note, in order to provide impacts (Donovan et al., 2014).
robust support for potential biomarkers of CTE, we lay
emphasis on studies that included populations presum- CORTICAL ATROPHY AND THINNING
ably at high risk for CTE, such as former (i.e., nonactive) High resolution T1 volumetric MRI may help to identify
professional contact sport athletes. Comprehensive p-τ-related neuronal loss in CTE. In the Gardner et al.
reviews of neuroimaging findings in the setting of acute (2015) case series study, seven of the nine former
and chronic RHI are provided elsewhere (Ng et al., 2014; American professional football players exhibited cortical
Koerte et al., 2015b). atrophy to a greater proportion than expected for their
age. Shrinkage and thinning of frontotemporal regions
Structural neuroimaging and aspects of the MTL (e.g., hippocampus) is likely
in CTE given these brain regions are affected by p-τ in
CAVUM SEPTUM PELLUCIDUM
the early stages of CTE and modulate clinical functions
A cavum septum pellucidum (CSP) is a consistent neu- impaired in CTE (e.g., episodic memory impairment,
ropathologic feature of CTE and represents a deviation executive dysfunction, depression, aggression, impulsiv-
of the embryologic fluid space between the leaflets of ity). Most compelling evidence supporting this notion
the septum pellucidum. A CSP can be observed on con- can be found in a recent study by Goswami et al.
ventional MRI. A recent study of 72 former NFL players (2016). In that study, the uncinate fasciculus differ-
and 14 former professional noncontact sport athletes who entiated former professional football players with a
underwent high resolution 3T MRI showed that the for- history of multiple concussions from healthy controls
mer NFL players had a higher rate of CSP, greater CSP (Goswami et al., 2016). The former professional foot-
length, and greater ratio of CSP length to septum length ball players with a history of concussion manifested
(Koerte et al., 2016). Greater length of the CSP predicted thinner anterior temporal lobes, and there was an
worse performance on a list learning and word reading inverse correlation between orbitofrontal cortex thick-
task. High rates of CSP on MRI was also found in another ness with aggression and task errors on a go/no-go task
346 M.L. ALOSCO AND R.A. STERN
(suggestive of impulsivity). There was also a negative superior frontal lobes, bilateral medial temporal lobes,
correlation between axial diffusivity and error rates and the left parietal lobe. The SUVRs in these regions
and aggression, and elevated resting state functional were further associated with years of American football
connectivity between the anterior temporal lobe and play. However, tau deposition did not correspond to cog-
the orbitofrontal cortex. Former NFL players have also nitive or neuropsychiatric tests. Notably, only 1 of the
been shown to exhibit greater right hippocampal atrophy 31 former NFL players had a positive florbetapir PET.
relative to age-matched healthy controls (Coughlin et al., The PET ligand 18F-FDDNP also targets p-τ, but
2015). Singh et al. (2014) also found that college football unlike 18F-AV1451 (and other similar recent ligands)
18
players with and without a history of concussion exhib- F-FDDNP is an old ligand that binds to both p-τ and
ited smaller hippocampal volumes relative to healthy b-amyloid, as well as other proteins, and thus lacks spe-
controls, and there was an inverse relationship between cific diagnostic accuracy. Nevertheless, Small et al.
years of football played and left hippocampal volume. (2013) found that FDDNP-PET signals were higher in
Players with a concussion had a smaller hippocampus a sample of five symptomatic former NFL players rela-
than those without such history. tive to demographically matched controls, specifically
in multiple subcortical regions and the amygdala. That
Functional neuroimaging same group reported 18F-FDDNP PET results in former
American professional football players that the authors
NEGATIVE AMYLOID AND POSITIVE TAU
(Barrio et al., 2015) stated were consistent with CTE
PET imaging is a nuclear medicine functional neuroim- neuropathology, though the pattern of findings was not
aging technique that detects g rays emitted from radio- consistent with those described by McKee et al. (2013,
labeled tracers injected into the body, which are 2015). Finally, PET ligands that target TBI-related
captured in a PET scanner and the data used to construct neuroinflammation (e.g., translocator protein, TSPO)
three-dimensional images. It is often used to examine may also have clinical utility in CTE (Coughlin
brain tissue metabolic activity using 18Fluorodeoxyglu- et al., 2015).
cose (FDG) as a tracer of glucose utilization. Three
PET ligands that bind to b-amyloid have recently been
HYPOMETABOLISM AND IMPAIRED NEURAL ACTIVITY
developed and approved by the Food and Drug Admin-
istration (e.g., 18F-Florbetapir) (Landau et al., 2013), and FDG-PET and other neuroimaging methods [e.g., arterial
used as part of the diagnostic process for AD, as well as spin labeling (ASL), single photon emission computed
for the detection of preclinical AD (Beach et al., 2014; tomography (SPECT), and functional MRI (fMRI)]
Pietrzak et al., 2015). Minimal to no uptake of the amy- examine cerebral blood flow (CBF) and metabolism pat-
loid tracer would rule out AD as an etiology. Given that terns that are distinct across neurodegenerative disorders.
b-amyloid is not a consistent feature of CTE, amyloid These tools may be useful to detect unique patterns of
PET imaging may be key for the differential diagnosis metabolic abnormalities in CTE. Peskind et al. (2011)
between CTE and AD in vivo; that is, absence of found FDG-PET hypometabolism in the MTL, cerebel-
b-amyloid plaques (as determined by negative amyloid lum, vermis, and pons in a sample of Iraq war veterans
PET) would be suggestive of CTE in a symptomatic indi- exposed to repeated blast exposures. In another study
vidual who does not meet diagnostic criteria for another of war veterans, repeated blast exposure subjects exhib-
disease or disorder. In recent years, several groups ited greater hypometabolism in the right superior parietal
have been developing ligands that bind to p-τ for use lobe relative to subjects exposed to blunt injury (Mendez
in humans (Harada et al., 2016). The PET ligand et al., 2013). ASL-related findings in cognitively
18
F-AV1451 has an affinity to bind to p-τ selectively over impaired former NFL players relative to controls also
b-amyloid and may have key diagnostic utility in tauopa- showed regional blood flow differences in the left
thies, such as CTE (Gandy and DeKosky, 2014). As an temporal pole, inferior parietal lobule, and superior
example, a case study found negative 18F-florbetapir temporal gyrus that corresponded to neuropsychologic
and positive 18F-AV1451 PET imaging in a former test performance (Hart et al., 2013).
NFL player who had a clinical profile similar to AD, Another useful fMRI biomarker is blood-oxygen-
but the combination of these biomarker findings ruled level dependent (BOLD) fMRI, which indirectly exam-
out AD as the likely etiology (Mitsis et al., 2014). Flor- ines neural activity through measurement of changes in
tauciper and florbetapir PET were recently examined in a CBF via contrast between oxygenated and deoxygenated
sample of 26 symptomatic former NFL players and blood. BOLD fMRI findings in a sample of former NFL
31 same-age asymptomatic controls without a history players has raised the possibility that recurrent concus-
of TBI or contact sport participation (Stern et al., sions may be associated with later-life functional ineffi-
2019). On average, flortaucipir SUVR was higher in for- ciencies in relational memory neural networks (Ford
mer NFL players compared to the controls in the bilateral et al., 2013). fMRI could be an important tool to better
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 347
understand the neural substrates of clinical impairment in and elevated CSF p-τ/τ ratio would theoretically be bio-
CTE, and much more research that uses fMRI in living markers of CTE. There is a paucity, however, of in vivo
subjects at high risk for CTE is needed. research examining whether fluid biomarkers can accu-
rately detect the long-term consequences of RHI expo-
NEUROCHEMICAL ALTERATIONS sure, in general, or underlying CTE pathology, in
particular. Recent findings from Stern et al. (2016) sup-
Magnetic resonance spectroscopy (MRS) is a safe,
port plasma exosomal tau as a potential biomarker for
nonradioactive, advanced neuroimaging method that
CTE. Exosomes are nanovesicles released by cells
examines physiologic metabolism in vivo. MRS mea-
throughout the body into the extracellular environment.
sures neurometabolic markers indicative of neuronal loss
Notably, they cross the blood–brain barrier and can be
(N-acetyl aspartate; NAA), excitotoxicity, gliosis, neu-
isolated in blood. Exosomes carry the molecular cargo
roinflammation (glutamate-glutamine; Glx, myo-
of the cell from which it was derived, including tau, if
inositol; mI), and axonal injury (choline; Cho) (Lin
present. Plasma and serum exosomal p-τ and total tau
et al., 2012; Ng et al., 2014), all of which are common
have both been shown to accurately detect AD
neuropathologic changes in CTE. Neurochemistry in
(Fiandaca et al., 2015). Stern et al. (2016) found that a
the posterior cingulate gyrus (PCG) was compared
sample of 78 former NFL players exhibited higher levels
between five former professional male athletes (three
of plasma exosomal tau relative to 16 controls without RHI
of five being former NFL players) with RHI exposure
exposure. Exosomal tau discriminated the former NFL
and five healthy controls without a history of RHI (Lin
players from controls with 82% sensitivity, and 100%
et al., 2015). All professional male athletes were exam-
specificity, and greater levels of exosomal tau in the
ined 3–25 years after their career and exhibited symp-
NFL group correlated with worse performance on neurop-
toms including, but not limited to, memory loss,
sychologic tests of memory and psychomotor speed. The
impaired judgment, aggression, and depression. Relative
authors acknowledge that their findings are preliminary
to controls, RHI exposure was associated with 31%
and require refinement of the procedures, as well as repli-
higher Glx and 65% higher Cho levels in the PCG. In
cation and validation. Alosco et al. also found an associa-
a follow-up MRS study of 77 symptomatic former
tion between greater exposure to RHI (using a cumulative
NFL players and 23 asymptomatic controls without a
head impact index) and higher levels of both plasma
history of TBI or contact sport participation (Alosco
(Alosco et al., 2016) and CSF (Alosco et al., 2018a,b) total
et al., 2019), there was lower NAA in the parietal white
tau in a sample of symptomatic former NFL players. In
matter among the former NFL players compared to con-
both studies, however, the fluid levels of total tau in the
trols. Exposure to RHI (estimated by a cumulative head
former NFL players were not significantly different from
impact index) was associated with lower creatinine in the
a group of aysmptomatic same-age individuals without a
parietal white matter of the former NFL players.
history of TBI or contact sport participation.
Neurochemicals in the anterior cingulate gyrus that are
associated with neuroinflammatory processes were also
RISK AND PROTECTIVE FACTORS
found to affect behavioral and mood functioning.
Other work has used MRS to examine neurochemical As previously highlighted, neuropathologic evidence
concentrations in 11 former professional soccer players supports RHI as a necessary risk factor for CTE diagno-
exposed to subconcussive trauma aged between 40 and sis, and we present research below that has examined
70 years and 14 age- and gender-matched noncontact RHI exposure and later-life neurologic impairment in
former athletes. Former soccer players exhibited higher living subjects. Because RHI exposure history alone is
Cho and mI concentrations in the PCG relative to con- not sufficient for the development of CTE, other risk
trols (Koerte et al., 2015a). Relative to former collegiate factors are believed to interact with RHI to either (1) con-
athletes without a history of concussion, those with a tribute to the development of CTE, and/or (2) contribute
concussion history exhibit elevated mI in the medial tem- to the symptom heterogeneity observed in CTE. Despite
poral lobe that correlated with worse visual memory the advancements of our understanding of the disease,
performance (Tremblay et al., 2013). CTE risk factors remain poorly understood, and there
is limited research on this topic. A nonexhaustive list
Fluid biomarkers of several potential targets is discussed later.
Fluid biomarkers are cost and time efficient alternatives
RHI exposure factors
to neuroimaging. Fluid biomarkers are typically derived
from blood or CSF. For example, CSF levels of b-amy- Rapidly expanding research has sought to quantify RHI
loid, p-τ, and total tau are known biomarkers of AD exposure in living subjects, in order to examine directly
(Kang et al., 2015; Olsson et al., 2016). Given the nature the relationship between RHI and later-life neurologic
of CTE neuropathology, normal b-amyloid CSF levels consequences. Quantification of RHI exposure history
348 M.L. ALOSCO AND R.A. STERN
in living subjects is challenging because it involves maturation (e.g., Caviness et al., 1996; Epstein, 1999;
retrospective self-report of numerous ambiguous events Giedd, 2008). It is hypothesized that RHI exposure dur-
throughout one’s life. Research has relied on single, indi- ing this window of neurodevelopmental vulnerability
rect metrics of RHI (e.g., recurrent concussion history). may disrupt normal brain development or result in a
These metrics have been shown to be associated with lower threshold for later life cognitive and structural
later-life clinical impairment (Guskiewicz et al., 2005, brain pathology associated with CTE. Notably, research
2007; Wright et al., 2016), but do not account for subcon- in active football players suggests that the effects of RHI
cussive events, perhaps the most important risk factor from youth football may not be observed immediately or
for CTE. even in young adulthood (Caccese et al., 2019) and thus
Direct metrics that reliably quantify both concussive there appears to be a possible interaction with aging.
and subconcussive trauma are beginning to be devel- Other aspects of RHI exposure (e.g., specific types of
oped. Montenigro et al. (2016) recently created the hits, such as rotational forces), the amount of rest
cumulative head impact index (CHII), a metric of cumu- between hits may also be key in the pathogenesis of
lative exposure to RHI from football. The CHII is based CTE, and this awaits empirical testing.
on self-reported exposure and estimated quantitation of
head impacts from published helmet accelerometer stud- Non-RHI exposure factors
ies. The CHII has been used to examine the relationship
between cumulative RHI exposure and later-life cogni- GENETICS
tive, mood, and behavioral impairment in a sample of McKee et al. (2013) found that, of the 68 contact sport
former football players whose highest level of play athletes and military veterans neuropathologically diag-
was either high school or college. The mean CHII was nosed with CTE, the proportion of those with at least one
7742 total impacts, and it strongly predicted later-life APOE E4 allele was similar to the general population. In
clinical outcomes and outperformed other individual contrast, Stern et al. (2013) found that relative to an
metrics such as concussion history. There was also a age-matched normative sample, there was a greater pres-
threshold of head impacts necessary for risk of cognitive ence of APOE genotypes in their sample of contact
or neurobehavioral impairment. In addition to the CHII, sport athletes with neuropathologically diagnosed CTE
Wright et al. (2016) recently developed an index and no other neuropathologic finding. There was a larger
comprised of concussion frequency, severity, and time- proportion of E4 homozygotes in the CTE subjects with
frame, as well as metrics of cognitive reserve, and found cognitive impairment as the initial presenting symptom,
that this index successfully predicted global cognitive and not the behavioral/mood subgroup. Possession of
ability in former professional American football players. an APOE E4 allele has also been shown to exacerbate
However, this index did not include estimates of subcon- clinical impairment in high exposure professional boxers
cussive brain trauma, among other limitations. Kerr et al. (i.e., 12 or more professional bouts) (Jordan et al., 1997),
(2015) proposed the Head Impact Exposure Estimate professional football players (Kutner et al., 2000), and is
(HIEE), which is a self-report estimate of a football associated with increased risk for dementia following a
player’s total hours of contact exposure. The relationship TBI (Mayeux et al., 1995). The APOE E4 allele is the
between HIEE and outcomes has yet to be determined most important genetic risk factor for AD, and future
and that metric does not include helmet accelerometer work will continue to examine its role in CTE, in addition
intensity estimates. to other genes, such as the microtubule-associated
Early age of first exposure (AFE) to tackle football, or protein tau (MAPT) gene.
RHI in general, may be involved in later risk for CTE.
Recent findings show that former NFL players who
CEREBROVASCULAR DISEASE
began playing tackle football prior to age 12 exhibited
worse cognitive functioning (Stamm et al., 2015a) and Cerebrovascular disease may interact with RHI to
microstructural integrity of the anterior corpus callosum contribute to the development of CTE. Pathologic corre-
(Stamm et al., 2015b) in middle age compared to those lates of white matter signal abnormalities (WMSA) seen
who began playing at age 12 or older. Among autopsy on MRI (e.g., gliosis, axonal injury) are common and
participants with neuropathologically confirmed CTE, persisting in former football and soccer players with
AFE to football was not associated with CTE pathologic extensive RHI exposure, and may contribute to memory
severity (Alosco et al., 2018a,b). However, younger AFE loss, executive dysfunction, aggression, and depression
was associated with earlier onset of cognitive and beha- (Lin et al., 2015; Koerte et al., 2015a). Concussion
vioral/mood symptoms. In boys, the years between the has been linked with cerebral hypoperfusion that may
ages of 9 and 12 represent a period of time in which persist (Wang et al., 2016), and boxers exhibit chronic
the brain undergoes tremendous neurodevelopmental frontotemporal cerebral hemodynamic impairment
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 349
(Bailey et al., 2013). The case series study by Gardner anabolic steroid use is prevalent in contact sport athletes,
et al. (2015) found that seven of nine former NFL players but has been shown to be unrelated to CTE neuropatho-
had subcortical or periventricular WMSA. In addition to logic severity (McKee et al., 2013). Moreover, many
RHI-related risk for WMSA, cardiovascular disease former American football players with neuropathologi-
(CVD) and its risk factors are prevalent in active and cally confirmed CTE played well before the availability
former professional American football players (Tucker and use of anabolic steroids or other performance
et al., 2009; Pokharel et al., 2014; Stamm et al., enhancing drugs. However, steroids are known to exac-
2015a), and is a common cause of death within this pop- erbate neuropsychiatric symptoms, and a recent mouse
ulation (Lehman et al., 2012). CVD risk factors (e.g., model study showed that repetitive TBI was associated
obesity) in former NFL players can reduce prefrontal with axonal injury and microgliosis, and that these
and temporal perfusion, which may underpin executive pathophysiologic changes were exacerbated by andro-
and memory deficits (Willeumier et al., 2012). genic–anabolic steroids (Namjoshi et al., 2016).

AGE CONCLUSIONS AND FUTURE


Older age at death is associated with greater CTE patho- DIRECTIONS
logic severity (McKee et al., 2013). Those who develop Chronic traumatic encephalopathy is a unique neurode-
cognitive impairment as an initial presenting symptom generative disease associated with a history of exposure
are typically older and have an advanced stage of CTE to RHI. Neuropathologic and clinical research in CTE
(Stern et al., 2013). There is also neuropathologic evi- has evolved rapidly over the past decade and has led to
dence of CTE in some younger individuals (e.g., in their improved understanding of this progressive and devas-
20s) who generally had predominantly behavioral/mood tating, but potentially preventable, brain disease. In par-
symptoms, and died with early stage CTE because of ticular, neuropathologic diagnostic criteria have been
suicide, overdose, or incidental death. This subgroup developed and clinical research criteria for the in vivo
of individuals may have developed cognitive impairment diagnosis of CTE have been proposed. The existing lit-
and more advanced CTE if they lived longer. In CTE, erature, however, is not without methodological limita-
age may be a modifier of disease progression vs a tions, and there remain many questions and knowledge
pathogenetic mechanism. gaps that need to be addressed. CTE research is of high
priority given the millions of individuals who could be
COGNITIVE RESERVE potentially impacted by this disease.
High cognitive reserve (e.g., as measured by greater The majority of existing neuropathologic CTE
years of education, high occupational attainment) is a research is limited by ascertainment bias due to samples
known clinical modifier of neuropathology in neuro- consisting of brain donors with extreme RHI exposure
degeneration (Stern, 2012), and there is strong prelimi- (e.g., professional contact sport athletes) and suspected
nary evidence for its protective effects in CTE. For pathology, and who themselves or their family members
example, in the examination of the clinical presentation have agreed to brain donation. It is important to note that
of neuropathologically confirmed cases of CTE without the objective of current postmortem CTE research is to
comorbid disease by Stern et al. (2013), two of three neuropathologically define CTE and its clinicopatho-
asymptomatic subjects who had stage II CTE neuropa- logic correlates and not to determine epidemiology;
thology also had advanced graduate degrees. Recent data thus, recruitment of subjects at high risk for CTE is
also supports a cognitive reserve effect (operationalized essential. Although the study from the Mayo Clinic’s
by academic achievement and occupational attainment) large, unbiased brain bank (Bieniek et al., 2015), pro-
in a sample of former professional American football vides some initial evidence of the prevalence of CTE
players (Wright et al., 2016). Higher cognitive reserve neuropathology in individuals with a history of amateur
has also been shown to attenuate cognitive dysfunction contact sports, large epidemiologic postmortem studies
and facilitate recovery following TBI (Kesler et al., are needed to determine the association between RHI
2003; Schneider et al., 2014). and CTE in former athletes, military veterans exposed
to blasts, as well as in nonathletic exposure to RHI
(e.g., work-related head trauma, domestic violence, falls
LIFESTYLE FACTORS
in elderly). Likewise, to date, nearly all neuropathologi-
Poor diet, lack of physical activity, and substance abuse cally diagnosed cases of CTE have been former male
behaviors can all increase vulnerability to neurode- football players. It will be important to examine larger
generation, although the specific contribution of these numbers of brains of other contact sport athletes exposed
factors to CTE remains unknown. Of note, a history of to RHI, particularly former female athletes.
350 M.L. ALOSCO AND R.A. STERN
Refinement of the existing neuropathologic criteria extraction) at baseline and again at a 3-year follow-up.
for CTE will be important for ex vivo differential In addition to biomarkers, DIAGNOSE CTE will refine
diagnosis from other neurodegenerative tauopathies. and validate clinical diagnostic criteria for CTE, and it
Postmortem studies that implement control and neurode- will examine various risk factors (e.g., genetics, life-
generative comparison groups will shed light on the style) that interact with RHI to contribute to the patho-
following: (1) the mechanisms of the spread of tau and genesis of CTE. Moreover, many of the subjects from
other proteins (e.g., b amyloid, TDP-43) involved in DIAGNOSE CTE will likely agree to brain donation,
CTE; (2) characterization of the extent and nature of which will generate research that can clarify the patho-
other pathologic aspects of CTE, including neuronal cell genetic mechanisms by which the acute neurologic
loss, gliosis, and inflammation; (3) identification of the consequences of RHI transition to CTE. Simultaneous
involvement of other cortical and subcortical brain to DIAGNOSE CTE, research from UNITE will con-
regions involved in the pathogenesis of CTE; and (4) tinue to conduct clinicopathologic studies in subjects
whether RHI exposure and CTE pathology are associated with neuropathologically confirmed CTE that will help
with an altered and accelerated development of a variety to determine the pathologic differences in symptom pre-
of neurodegenerations. Validation of the McKee et al. sentation (e.g., behavior/mood symptoms vs cognitive
(2013) staging classification for the severity and progres- impairment). Once CTE can be diagnosed during life
sion of p-τ in CTE is also a target of future work. and risk factors are identified, prevalence and incidence
The critical next step is to be able to diagnose CTE of this disease can be determined, therapeutic targets can
during life, in order to facilitate clinical research on the be identified or developed, clinical trials can be initiated,
nature and course of symptoms, epidemiology, risk fac- and clinical and policy-related issues can be addressed.
tors, and disease-specific mechanisms. Currently, the
diagnosis of “Probable CTE” is limited due to the lack REFERENCES
of validated biomarkers. This is problematic because
the clinical manifestations of CTE are somewhat similar Abbas K, Shenk TE, Poole VN et al. (2015). Effects of repet-
to other neurodegenerative diseases (e.g., AD, FTD), and itive sub-concussive brain injury on the functional connec-
tivity of Default Mode Network in high school football
there is much symptom overlap with psychiatric disor-
athletes. Dev Neuropsychol 40: 51–56.
ders (e.g., posttraumatic stress disorder, depression), Alosco ML, Tripodis Y, Jarnagin J et al. (2016). Repetitive
notably for CTE cases with predominantly behavioral/ head impact exposure and later-life plasma total tau in
mood symptoms. In addition, clinically differentiating former National Football League players. Alzheimers
between CTE and persistent postconcussive syndrome Dement 7: 33–40.
among young individuals with active or recently con- Alosco ML, Mez J, Tripodis Y et al. (2018a). Age of first
cluded RHI exposure is challenging, as highlighted by exposure to tackle football and chronic traumatic enceph-
a recent published case report (Mez et al., 2016). alopathy. Ann Neurol 83: 886–901.
A grant was recently funded by NINDS for a 7-year, Alosco ML, Tripodis Y, Fritts NG et al. (2018b). Cerebrospinal
multicenter study, entitled, “Diagnostics, Imaging, fluid tau, Ab, and sTREM2 in Former National Football
and Genetics Network for the Objective Study and League players: modeling the relationship between repeti-
tive head impacts, microglial activation, and neurodegen-
Evaluation of Chronic Traumatic Encephalopathy
eration. Alzheimers Dement 14: 1159–1170.
(DIAGNOSE CTE) Research Project” (PIs: Robert A. Alosco ML, Tripodis Y, Rowland B et al. (2019). A magnetic res-
Stern [Contact PI], Jeffrey Cummings, Eric Reiman, onance spectroscopy investigation in symptomatic former
Martha Shenton). That study is currently being con- NFL players. Brain Imaging Behav (Epub ahead of print).
ducted with the primary objective of developing Armstrong RA, McKee AC, Stein TD et al. (2017).
identification of biomarkers that support the diagnosis A quantitative study of tau pathology in eleven cases of
of “Probable CTE” during life. DIAGNOSE CTE will chronic traumatic encephalopathy. Neuropathol Appl
include former athletes with high exposure to RHI Neurobiol 43: 154–166.
(former NFL players with and without symptoms), Bailes JE, Petraglia AL, Omalu BI et al. (2013). Role of
medium exposure to RHI (former college football subconcussion in repetitive mild traumatic brain injury.
players with and without CTE), and a control group J Neurosurg 119: 1235–1245.
Bailey DM, Jones DW, Sinnott A et al. (2013). Impaired
(asymptomatic same-age men without RHI exposure
cerebral haemodynamic function associated with chronic
or TBI). All subjects will undergo clinical examinations traumatic brain injury in professional boxers. Clin Sci
(neurologic, motor, neuropsychologic, neuropsychiat- (Lond) 124: 177–189.
ric, and functional assessments), extensive neuroimag- Barrio JR, Small GW, Wong KP et al. (2015). In vivo charac-
ing (amyloid and tau PET, MRI, fMRI, DTI, MRS), terization of chronic traumatic encephalopathy using
lumbar punctures (for CSF biomarkers), and blood [F-18]FDDNP PET brain imaging. Proc Natl Acad Sci
draws (for blood-based biomarkers, as well as DNA USA 112: E2039–E2047.
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 351
Baugh CM, Robbins CA, Stern RA et al. (2014). Current Cloots RJ, Gervaise HM, van Dommelen JA et al. (2008).
understanding of chronic traumatic encephalopathy. Curr Biomechanics of traumatic brain injury: influences of the
Treat Options Neurol 16: 306. morphologic heterogeneities of the cerebral cortex. Ann
Bazarian JJ, Zhu T, Zhong J et al. (2014). Persistent, long-term Biomed Eng 36: 1203–1215.
cerebral white matter changes after sports-related repetitive Cloots RJ, van Dommelen JA, Nyberg T et al. (2011).
head impacts. PLoS One 9: e94734. Micromechanics of diffuse axonal injury: influence of
Beach TG, Schneider JA, Sue LI et al. (2014). Theoretical axonal orientation and anisotropy. Biomech Model
impact of Florbetapir (18F) amyloid imaging on diagnosis Mechanobiol 10: 413–422.
of Alzheimer dementia and detection of preclinical cortical Collins MW, Lovell MR, Iverson GL et al. (2002). Cumulative
amyloid. J Neuropathol Exp Neurol 73: 948–953. effects of concussion in high school athletes. Neurosurgery
Beitz JM (2014). Parkinson’s disease: a review. Front Biosci 51: 1175–1179. discussion 1180–1171.
(Schol Ed) 6: 65–74. Corsellis JA, Bruton CJ, Freeman-Browne D (1973). The
Belanger HG, Vanderploeg RD (2005). The neuropsycholog- aftermath of boxing. Psychol Med 3: 270–303.
ical impact of sports-related concussion: a meta-analysis. Coughlin JM, Wang Y, Munro CA et al. (2015).
J Int Neuropsychol Soc 11: 345–357. Neuroinflammation and brain atrophy in former NFL
Belanger HG, Curtiss G, Demery JA et al. (2005). Factors players: an in vivo multimodal imaging pilot study.
moderating neuropsychological outcomes following mild Neurobiol Dis 74: 58–65.
traumatic brain injury: a meta-analysis. J Int Crane PK, Gibbons LE, Dams-O’Connor K et al. (2016).
Neuropsychol Soc 11: 215–227. Association of traumatic brain injury with late-life neuro-
Belanger HG, Vanderploeg RD, McAllister T (2015). degenerative conditions and neuropathologic findings.
Subconcussive blows to the head: a formative review of JAMA Neurol 73: 1062–1069.
short-term clinical outcomes. J Head Trauma Rehabil 31: Crisco JJ, Fiore R, Beckwith JG et al. (2010). Frequency and
159–166. location of head impact exposures in individual collegiate
Bieniek KF, Ross OA, Cormier KA et al. (2015). Chronic trau- football players. J Athl Train 45: 549–559.
matic encephalopathy pathology in a neurodegenerative Daneshvar DH, Nowinski CJ, McKee AC et al. (2011). The
disorders brain bank. Acta Neuropathol 130: 877–889. epidemiology of sport-related concussion. Clin Sports
Bigler ED (2008). Neuropsychology and clinical neuroscience Med 30: 1–17. vii.
of persistent post-concussive syndrome. J Int Neuropsychol Dashnaw ML, Petraglia AL, Bailes JE (2012). An overview of
Soc 14: 1–22. the basic science of concussion and subconcussion: where we
Blaylock RL, Maroon J (2011). Immunoexcitotoxicity as a are and where we are going. Neurosurg Focus 33. E5: 1–9.
central mechanism in chronic traumatic encephalopathy- Davenport EM, Whitlow CT, Urban JE et al. (2014). Abnormal
A unifying hypothesis. Surg Neurol Int 2: 107. white matter integrity related to head impact exposure in a
Blumbergs PC, Scott G, Manavis J et al. (1994). Staining of season of high school varsity football. J Neurotrauma 31:
amyloid precursor protein to study axonal damage in mild 1617–1624.
head injury. Lancet 344: 1055–1056. Defense and Veterans Brain Injury Center. (2015). DoD
Bowman KM, Blau A (1940). Psychotic states following head Worldwide Numbers for TBI. Retrieved from http://
and brain injury in adults and children. In: S Brock (Ed.), dvbic.dcoe.mil/dod-worldwide-numbers-tbi.
Injuries of the skull, brain and spinal cord: neuro- Dikmen S, Machamer J, Temkin N (2001). Mild head injury:
psychiatric, surgical, and medico-legal aspects. facts and artifacts. J Clin Exp Neuropsychol 23: 729–738.
Williams & Wilkins Co, Baltimore, MD. Donovan V, Kim C, Anugerah AK et al. (2014). Repeated mild
Brandenburg W, Hallervorden J (1954). Dementia pugilistica traumatic brain injury results in long-term white-matter
with anatomical findings. Virchows Arch 325: 680–709. disruption. J Cereb Blood Flow Metab 34: 715–723.
Broglio SP, Eckner JT, Martini D et al. (2011). Cumulative head Epstein HT (1999). Stages of increased cerebral blood flow
impact burden in high school football. J Neurotrauma 28: accompany stages of rapid brain growth. Brain Dev 21:
2069–2078. 535–539.
Caccese JB, DeWolf RM, Kaminski TW et al. (2019). Erlanger DM (2015). Exposure to sub-concussive head injury
Estimated age of first exposure to American football and in boxing and other sports. Brain Inj 29: 171–174.
neurocognitive performance amongst NCAA male Fainaru-Wada M, Fainaru S (2013). League of denial: the
student-athletes: a cohort study. Sports Med49: 477–487. NFL, concussions and the battle for truth, Crown
https://doi.org/10.1007/s40279-019-01069-x. Archetype, New York.
Carroll Jr EJ (1936). Punch-drunk. Am J Med Sci 191: Falcon B, Zivanov J, Zhang W et al. (2019). Novel tau filament
706–712. fold in chronic traumatic encephalopathy encloses hydro-
Caviness Jr VS, Kennedy DN, Richelme C et al. (1996). The phobic molecules. Nature568: 420–423. https://doi.org/
human brain age 7–11 years: a volumetric analysis based 10.1038/s41586-019-1026-5.
on magnetic resonance images. Cereb Cortex 6: 726–736. Fiandaca MS, Kapogiannis D, Mapstone M et al. (2015).
Chappell MH, Ulug AM, Zhang L et al. (2006). Distribution of Identification of preclinical Alzheimer’s disease by a
microstructural damage in the brains of professional profile of pathogenic proteins in neurally derived blood
boxers: a diffusion MRI study. J Magn Reson Imaging exosomes: a case-control study. Alzheimers Dement 11:
24: 537–542. 600–607. e601.
352 M.L. ALOSCO AND R.A. STERN
Fleminger S, Oliver DL, Lovestone S et al. (2003). Head injury Iverson GL (2005). Outcome from mild traumatic brain injury.
as a risk factor for Alzheimer’s disease: the evidence Curr Opin Psychiatry 18: 301–317.
10 years on; a partial replication. J Neurol Neurosurg Iverson GL, Gaetz M, Lovell MR et al. (2004). Cumulative effects
Psychiatry 74: 857–862. of concussion in amateur athletes. Brain Inj 18: 433–443.
Ford JH, Giovanello KS, Guskiewicz KM (2013). Episodic Jack Jr CR, Albert MS, Knopman DS et al. (2011).
memory in former professional football players with a Introduction to the recommendations from the National
history of concussion: an event-related functional neuroim- Institute on Aging-Alzheimer’s Association workgroups
aging study. J Neurotrauma 30: 1683–1701. on diagnostic guidelines for Alzheimer’s disease.
Gaetz M (2004). The neurophysiology of brain injury. Clin Alzheimers Dement 7: 257–262.
Neurophysiol 115: 4–18. Jellinger KA (2012). Interaction between pathogenic proteins
Gandy S, DeKosky ST (2014). [18F]-T807 tauopathy PET imag- in neurodegenerative disorders. J Cell Mol Med 16:
ing in chronic traumatic encephalopathy. F1000Res 3: 229. 1166–1183.
Gardner RC, Possin KL, Hess CP et al. (2015). Evaluating Johnson VE, Stewart W, Smith DH (2012). Widespread tau
and treating neurobehavioral symptoms in professional and amyloid-beta pathology many years after a single trau-
American football players: lessons from a case series. matic brain injury in humans. Brain Pathol 22: 142–149.
Neurol Clin Pract 5: 285–295. Jordan BD (2013). The clinical spectrum of sport-related
Gardner RC, Hess CP, Brus-Ramer M et al. (2016). Cavum traumatic brain injury. Nat Rev Neurol 9: 222–230.
septum pellucidum in retired American pro-football Jordan BD, Relkin NR, Ravdin LD et al. (1997).
players. J Neurotrauma 33: 157–161. Apolipoprotein E epsilon4 associated with chronic trau-
Giedd JN (2008). The teen brain: insights from neuroimaging. matic brain injury in boxing. JAMA 278: 136–140.
J Adolesc Health 42: 335–343. Kang JH, Korecka M, Figurski MJ et al. (2015). The Alzheimer’s
Giza CC, Hovda DA (2014). The new neurometabolic cascade disease neuroimaging initiative 2 biomarker core: a review of
of concussion. Neurosurgery 75: S24–S33. progress and plans. Alzheimers Dement 11: 772–791.
Goldstein LE, Fisher AM, Tagge CA et al. (2012). Chronic Katz RT, DeLuca J (1992). Sequelae of minor traumatic brain
traumatic encephalopathy in blast-exposed military vet- injury. Am Fam Physician 46: 1491–1498.
erans and a blast neurotrauma mouse model. Sci Transl Kay T, Harrington DE, Adams R et al. (1993). Definition of mild
Med 4: 134ra160. traumatic brain injury. J Head Trauma Rehabil 8: 86–87.
Goldstein LE, McKee AC, Stanton PK (2014). Considerations Kerr ZY, Littleton AC, Cox LM et al. (2015). Estimating con-
for animal models of blast-related traumatic brain injury tact exposure in football using the head impact exposure
and chronic traumatic encephalopathy. Alzheimers Res estimate. J Neurotrauma 32: 1083–1089.
Ther 6: 64. Kesler SR, Adams HF, Blasey CM et al. (2003). Premorbid
Goswami R, Dufort P, Tartaglia MC et al. (2016). intellectual functioning, education, and brain size in trau-
Frontotemporal correlates of impulsivity and machine matic brain injury: an investigation of the cognitive reserve
learning in retired professional athletes with a history of hypothesis. Appl Neuropsychol 10: 153–162.
multiple concussions. Brain Struct Funct 221: 1911–1925 Koerte IK, Kaufmann D, Hartl E et al. (2012a). A prospective
[Epub ahead of print]. study of physician-observed concussion during a varsity
Guskiewicz KM, McCrea M, Marshall SW et al. (2003). university hockey season: white matter integrity in ice
Cumulative effects associated with recurrent concussion hockey players. Part 3 of 4. Neurosurg Focus 6: 1–7.
in collegiate football players: the NCAA Concussion Koerte IK, Ertl-Wagner B, Reiser M et al. (2012b). White
Study. JAMA 290: 2549–2555. matter integrity in the brains of professional soccer players
Guskiewicz KM, Marshall SW, Bailes J et al. (2005). without a symptomatic concussion. JAMA 308: 1859–1861.
Association between recurrent concussion and late-life Koerte IK, Lin AP, Muehlmann M et al. (2015a). Altered
cognitive impairment in retired professional football neurochemistry in former professional soccer players with-
players. Neurosurgery 57: 719–726 (discussion 719–726). out a history of concussion. J Neurotrauma 32: 1287–1293.
Guskiewicz KM, Marshall SW, Bailes J et al. (2007). Recurrent Koerte IK, Lin AP, Willems A et al. (2015b). A review of
concussion and risk of depression in retired professional neuroimaging findings in repetitive brain trauma. Brain
football players. Med Sci Sports Exerc 39: 903–909. Pathol 25: 318–349.
Gysland SM, Mihalik JP, Register-Mihalik JK et al. (2012). Koerte IK, Hufschmidt J, Muehlmann M et al. (2016). Cavum
The relationship between subconcussive impacts and con- septi pellucidi in symptomatic former professional football
cussion history on clinical measures of neurologic function players. J Neurotrauma 33: 346–353.
in collegiate football players. Ann Biomed Eng 40: 14–22. Kutner KC, Erlanger DM, Tsai J et al. (2000). Lower cognitive
Harada R, Okamura N, Furumoto S et al. (2016). performance of older football players possessing apolipo-
Characteristics of tau and its ligands in PET imaging. protein E epsilon4. Neurosurgery 47: 651–657 (discussion
Biomolecules 6: 7. 657–658).
Hart Jr J, Kraut MA, Womack KB et al. (2013). Neuroimaging Landau SM, Breault C, Joshi AD et al. (2013). Amyloid-beta
of cognitive dysfunction and depression in aging retired imaging with Pittsburgh compound B and florbetapir: com-
National Football League players: a cross-sectional study. paring radiotracers and quantification methods. J Nucl Med
JAMA Neurol 70: 326–335. 54: 70–77.
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 353
Langlois JA, Rutland-Brown W, Wald MM (2006). The epi- McKee AC, Daneshvar DH, Alvarez VE et al. (2014). The
demiology and impact of traumatic brain injury: a brief neuropathology of sport. Acta Neuropathol 127: 29–51.
overview. J Head Trauma Rehabil 21: 375–378. McKee AC, Stein TD, Kiernan PT et al. (2015). The neuropa-
Lee EB, Kinch K, Johnson VE et al. (2019). Chronic traumatic thology of chronic traumatic encephalopathy. Brain Pathol
encephalopathy is a common co-morbidity, but less 25: 350–364.
frequent primary dementia in former soccer and rugby McKee AC, Cairns NJ, Dickson DW et al. (2016). The first
players. Acta Neuropathol138: 389–399. https://doi.org/ NINDS/NIBIB consensus meeting to define neuropatho-
10.1007/s00401-019-02030-y. logical criteria for the diagnosis of chronic traumatic
Lehman EJ, Hein MJ, Baron SL et al. (2012). encephalopathy. Acta Neuropathol 131: 75–86.
Neurodegenerative causes of death among retired National McKhann GM, Knopman DS, Chertkow H et al. (2011). The
Football League players. Neurology 79: 1970–1974. diagnosis of dementia due to Alzheimer’s disease:
Lin AP, Liao HJ, Merugumala SK et al. (2012). Metabolic recommendations from the National Institute on Aging-
imaging of mild traumatic brain injury. Brain Imaging Alzheimer’s Association workgroups on diagnostic guide-
Behav 6: 208–223. lines for Alzheimer’s disease. Alzheimers Dement 7:
Lin AP, Ramadan S, Stern RA et al. (2015). Changes in the 263–269.
neurochemistry of athletes with repetitive brain trauma: Medana IM, Esiri MM (2003). Axonal damage: a key
preliminary results using localized correlated spectros- predictor of outcome in human CNS diseases. Brain 126:
copy. Alzheimers Res Ther 7: 13. 515–530.
Ling H, Holton JL, Shaw K et al. (2015). Histological evidence Mendez MF, Owens EM, Reza Berenji G et al. (2013). Mild
of chronic traumatic encephalopathy in a large series traumatic brain injury from primary blast vs. blunt forces:
of neurodegenerative diseases. Acta Neuropathol 130: post-concussion consequences and functional neuroimag-
891–893. ing. NeuroRehabilitation 32: 397–407.
Ling H, Morris HR, Neal JW et al. (2017). Mixed pathologies Mez J, Solomon TM, Daneshvar DH et al. (2015). Assessing
including chronic traumatic encephalopathy account for clinicopathological correlation in chronic traumatic
dementia in retired association football (soccer) players. encephalopathy: rationale and methods for the UNITE
Acta Neuropathol133: 337–352. https://doi.org/10.1007/ study. Alzheimers Res Ther 7: 62.
s00401-017-1680-3. Mez J, Solomon TM, Daneshvar DH et al. (2016).
Lipton ML, Kim N, Zimmerman ME et al. (2013). Soccer Pathologically confirmed chronic traumatic encephalopa-
heading is associated with white matter microstructural thy in a 25-year-old former college football player.
and cognitive abnormalities. Radiology 268: 850–857. JAMA Neurol 73: 353–355.
Losoi H, Silverberg ND, W€aljas M et al. (2016). Recovery Mez J, Daneshvar DH, Kiernan PT et al. (2017).
from mild traumatic brain injury in previously healthy Clinicopathological evaluation of chronic traumatic enceph-
adults. J Neurotrauma 33: 766–776. alopathy in players of American football. JAMA318:
Mannix R, Berglass J, Berkner J et al. (2014). Chronic gliosis 360–370. https://doi.org/10.1001/jama.2017.8334.
and behavioral deficits in mice following repetitive mild Millspaugh JA (1937). Dementia pugilistica. US Naval Med
traumatic brain injury. J Neurosurg 121: 1342–1350. Bull 35: 297–303.
Martland HS (1928). Punch drunk. JAMA 91: 1103–1107. Mitsis EM, Riggio S, Kostakoglu L et al. (2014). Tauopathy
Martland HS (1943). Intracranial injuries and their sequelae PET and amyloid PET in the diagnosis of chronic traumatic
and punch drunk. Ther Intern Dis 3: 291–301. encephalopathies: studies of a retired NFL player and of a
Mawdsley C, Ferguson FR (1963). Neurological disease in man with FTD and a severe head injury. Transl Psychiatry
boxers. Lancet 2: 795–801. 4: e441.
Maxwell WL, Povlishock JT, Graham DL (1997). Montenigro PH, Baugh CM, Daneshvar DH et al. (2014).
A mechanistic analysis of nondisruptive axonal injury: a Clinical subtypes of chronic traumatic encephalopathy:
review. J Neurotrauma 14: 419–440. literature review and proposed research diagnostic criteria
Mayeux R, Ottman R, Maestre G et al. (1995). Synergistic for traumatic encephalopathy syndrome. Alzheimers Res
effects of traumatic head injury and apolipoprotein-epsilon Ther 6: 68.
4 in patients with Alzheimer’s disease. Neurology 45: Montenigro PH, Corp DT, Stein TD et al. (2015). Chronic
555–557. traumatic encephalopathy: historical origins and current
McCrory P, Meeuwisse WH, Aubry M et al. (2013). Consensus perspective. Annu Rev Clin Psychol 11: 309–330.
statement on concussion in sport: the 4th International Montenigro PH, Alosco ML, Martin B et al. (2016).
Conference on Concussion in Sport held in Zurich, Cumulative head impact exposure predicts later-life
November 2012. Br J Sports Med 47: 250–258. depression, apathy, executive dysfunction, and cognitive
McKee AC, Gavett BE, Stern RA et al. (2010). TDP-43 pro- impairment in former high school and college football
teinopathy and motor neuron disease in chronic traumatic players. J Neurotrauma 34: 328–340.
encephalopathy. J Neuropathol Exp Neurol 69: 918–929. Montine TJ, Phelps CH, Beach TG et al. (2012). National insti-
McKee AC, Stern RA, Nowinski CJ et al. (2013). The spec- tute on aging-Alzheimer’s Association guidelines for the
trum of disease in chronic traumatic encephalopathy. neuropathologic assessment of Alzheimer’s disease: a
Brain 136: 43–64. practical approach. Acta Neuropathol 123: 1–11.
354 M.L. ALOSCO AND R.A. STERN
Morales R, Green KM, Soto C (2009). Cross currents in protein Ponsford J, Willmott C, Rothwell A et al. (2000). Factors
misfolding disorders: interactions and therapy. CNS influencing outcome following mild traumatic brain injury
Neurol Disord Drug Targets 8: 363–371. in adults. J Int Neuropsychol Soc 6: 568–579.
Mouzon BC, Bachmeier C, Ferro A et al. (2014). Chronic Povlishock JT (1993). Pathobiology of traumatically induced
neuropathological and neurobehavioral changes in a repet- axonal injury in animals and man. Ann Emerg Med 22:
itive mild traumatic brain injury model. Ann Neurol 75: 980–986.
241–254. Sato T, Takeuchi S, Saito A et al. (2009). Axonal ligation
Namjoshi DR, Cheng WH, Carr M et al. (2016). Chronic expo- induces transient redistribution of TDP-43 in brainstem
sure to androgenic-anabolic steroids exacerbates axonal motor neurons. Neuroscience 164: 1565–1578.
injury and microgliosis in the CHIMERA mouse model Sayed N, Culver C, Dams-O’Connor K et al. (2013). Clinical
of repetitive concussion. PLoS One 11: e0146540. phenotype of dementia after traumatic brain injury.
Newell KL, Hyman BT, Growdon JH et al. (1999). Application J Neurotrauma 30: 1117–1122.
of the National Institute on Aging (NIA)-Reagan Institute Schneider EB, Sur S, Raymont V et al. (2014).
criteria for the neuropathological diagnosis of Alzheimer Functional recovery after moderate/severe traumatic
disease. J Neuropathol Exp Neurol 58: 1147–1155. brain injury: a role for cognitive reserve? Neurology 82:
Ng TS, Lin AP, Koerte IK et al. (2014). Neuroimaging in repet- 1636–1642.
itive brain trauma. Alzheimers Res Ther 6: 10. Scott G, Ramlackhansingh AF, Edison P et al. (2016).
Ojo JO, Mouzon B, Greenberg MB et al. (2013). Repetitive Amyloid pathology and axonal injury after brain trauma.
mild traumatic brain injury augments tau pathology and Neurology 86: 821–828.
glial activation in aged hTau mice. J Neuropathol Exp Seichepine DR, Stamm JM, Daneshvar DH et al. (2013).
Neurol 72: 137–151. Profile of self-reported problems with executive func-
Olsson B, Lautner R, Andreasson U et al. (2016). CSF and tioning in college and professional football players.
blood biomarkers for the diagnosis of Alzheimer’s disease: J Neurotrauma 30: 1299–1304.
a systematic review and meta-analysis. Lancet Neurol Apr Sephton CF, Cenik C, Kucukural A et al. (2011). Identification
8: 673–684 (Epub ahead of print). of neuronal RNA targets of TDP-43-containing ribonucleo-
Omalu BI, DeKosky ST, Minster RL et al. (2005). Chronic protein complexes. J Biol Chem 286: 1204–1215.
traumatic encephalopathy in a National Football League Sephton CF, Cenik B, Cenik BK et al. (2012). TDP-43 in
player. Neurosurgery 57: 128–134 (discussion 128–134). central nervous system development and function: clues
Omalu B, Fitzsimmons RP, Hammers J et al. (2010a). Chronic to TDP-43-associated neurodegeneration. Biol Chem
traumatic encephalopathy in a professional American 393: 589–594.
wrestler. J Forensic Nurs 6: 130–136. Shenton ME, Hamoda HM, Schneiderman JS et al. (2012).
Omalu BI, Hamilton RL, Kamboh MI et al. (2010b). Chronic A review of magnetic resonance imaging and diffusion ten-
traumatic encephalopathy in a National Football League sor imaging findings in mild traumatic brain injury. Brain
player: case report and emerging medicolegal practice Imaging Behav 6: 137–192.
questions. J Forensic Nurs 6: 40–46. Singh R, Meier TB, Kuplicki R et al. (2014). Relationship
Omalu BI, Hammers JL, Bailes J et al. (2011). Chronic trau- of collegiate football experience and concussion with
matic encephalopathy in an Iraqi war veteran with posttrau- hippocampal volume and cognitive outcomes. JAMA
matic stress disorder who committed suicide. Neurosurg 311: 1883–1888.
Focus 31: E3. Small GW, Kepe V, Siddarth P et al. (2013). PET scanning of
Oppenheimer DR (1968). Microscopic lesions in the brain brain tau in retired national football league players:
following head injury. J Neurol Neurosurg Psychiatry 31: preliminary findings. Am J Geriatr Psychiatry 21:
299–306. 138–144.
Osnato M, Giliberti V (1927). Postconcussion neurosis- Smith DH, Chen XH, Nonaka M et al. (1999). Accumulation of
traumatic encephalitis: a conception of postconcussion amyloid beta and tau and the formation of neurofilament
phenomena. Arch Neurol Psychiatr 18: 181–214. inclusions following diffuse brain injury in the pig.
Peskind ER, Petrie EC, Cross DJ et al. (2011). J Neuropathol Exp Neurol 58: 982–992.
Cerebrocerebellar hypometabolism associated with repeti- Smith DH, Johnson VE, Stewart W (2013). Chronic neuro-
tive blast exposure mild traumatic brain injury in 12 Iraq pathologies of single and repetitive TBI: substrates of
war Veterans with persistent post-concussive symptoms. dementia? Nat Rev Neurol 9: 211–221.
Neuroimage 54: S76–S82. Stamm JM, Bourlas AP, Baugh CM et al. (2015a). Age
Pietrzak RH, Lim YY, Neumeister A et al. (2015). Amyloid-ß, of first exposure to football and later-life cognitive
anxiety, and cognitive decline in preclinical Alzheimer impairment in former NFL players. Neurology 84:
disease: a multicenter, prospective cohort study. JAMA 1114–1120.
Psychiatry 72: 284–291. Stamm JM, Koerte IK, Muehlmann M et al. (2015b). Age at
Pokharel Y, Basra S, Lincoln AE et al. (2014). Association of first exposure to football is associated with altered
body mass index and waist circumference with subclinical corpus callosum white matter microstructure in former
atherosclerosis in retired NFL players. South Med J 107: professional football players. J Neurotrauma 32:
633–639. 1768–1776.
THE LONG-TERM CONSEQUENCES OF REPETITIVE HEAD IMPACTS 355
Stein TD, Alvarez VE, McKee AC (2014). Chronic traumatic Viano DC, Casson IR, Pellman EJ et al. (2005). Concussion
encephalopathy: a spectrum of neuropathological changes in professional football: comparison with boxing head
following repetitive brain trauma in athletes and military impacts—part 10. Neurosurgery 57: 1154–1172 (discus-
personnel. Alzheimers Res Ther 6: 4. sion 1154–1172).
Stein TD, Alvarez VE, McKee AC (2015a). Concussion in Victoroff J (2013). Traumatic encephalopathy: review and pro-
chronic traumatic encephalopathy. Curr Pain Headache visional research diagnostic criteria. NeuroRehabilitation
Rep 19: 47. 32: 211–224.
Stein TD, Montenigro PH, Alvarez VE et al. (2015b). Beta- W€aljas M, Iverson GL, Lange RT et al. (2015). A prospective
amyloid deposition in chronic traumatic encephalopathy. study of the persistent post-concussion symptoms fol-
Acta Neuropathol 130: 21–34. lowing mild traumatic brain injury. J Neurotrauma 32:
Stern Y (2012). Cognitive reserve in ageing and Alzheimer’s 534–547.
disease. Lancet Neurol 11: 1006–1012. Wang Y, Nelson LD, LaRoche AA et al. (2016). Cerebral
Stern RA, Daneshvar DH, Baugh CM et al. (2013). Clinical blood flow alterations in acute sport-related concussion.
presentation of chronic traumatic encephalopathy. J Neurotrauma 33: 1227–1236.
Neurology 81: 1122–1129. Weiner MW, Harvey D, Hayes J et al. (2017). Effects of
Stern RA, Tripodis Y, Baugh CM et al. (2016). Preliminary traumatic brain injury and posttraumatic stress disorder on
study of plasma exosomal tau as a potential biomarker development of Alzheimer’s disease in Vietnam Veterans
for chronic traumatic encephalopathy. J Alzheimers Dis using the Alzheimer’s disease neuroimaging initiative:
51: 1099–1109. preliminary report. Alzheimers Dement 3: 177–188.
Stern RA, Adler CH, Chen K et al. (2019). Tau positron- Willeumier K, Taylor DV, Amen DG (2012). Elevated body
emission tomography in former national football league mass in National Football League players linked to cogni-
players. N Engl J Med380: 1716–1725. https://doi.org/ tive impairment and decreased prefrontal cortex and tem-
10.1056/NEJMoa1900757. poral pole activity. Transl Psychiatry 2: e68.
Strain J, Didehbani N, Cullum CM et al. (2013). Depressive Wright MJ, Woo E, Birath JB et al. (2016). An index predictive
symptoms and white matter dysfunction in retired NFL of cognitive outcome in retired professional American
players with concussion history. Neurology 81: 25–32. Football players with a history of sports concussion.
Sugarman MA, McKee AC, Stein TD et al. (2019). Failure to J Clin Exp Neuropsychol 38: 561–571.
detect an association between self-reported traumatic brain Zhang L, Ravdin LD, Relkin N et al. (2003). Increased
injury and Alzheimer’s disease neuropathology and diffusion in the brain of professional boxers: a preclinical
dementia. Alzheimers Dement 15: 686–698. sign of traumatic brain injury? AJNR Am J Neuroradiol
Tagge CA, Fisher AM, Minaeva OV et al. (2018). Concussion, 24: 52–57.
microvascular injury, and early tauopathy in young athletes
after impact head injury and an impact concussion mouse FURTHER READING
model. Brain 141: 422–458.
Talavage TM, Nauman EA, Breedlove EL et al. (2014). CDC (2006). Behavioral risk factor surveillance system:
Functionally-detected cognitive impairment in high school exercise.
football players without clinically-diagnosed concussion. Center DaVBI (2015). DoD Worldwide Numbers for
J Neurotrauma 31: 327–338. TBI [Online], Available: http://dvbic.dcoe.mil/dod-
Tremblay S, De Beaumont L, Henry LC et al. (2013). Sports worldwide-numbers-tbi.
concussions and aging: a neuroimaging investigation. National Council of Youth Sports (2008). Report on Trends
Cereb Cortex 23: 1159–1166. and Participation in Organized Youth Sports, National
Tucker AM, Vogel RA, Lincoln AE et al. (2009). Prevalence Council of Youth Sports, Stuart, FL.
of cardiovascular disease risk factors among National Sports NCoY (2008). Report on trends and participation in
Football League players. JAMA 301: 2111–2119. organized youth sports.

You might also like