4,995
Views
68
CrossRef citations to date
0
Altmetric
Review Articles

Injury cascades in TBI-related neurodegeneration

& ORCID Icon
Pages 1177-1182 | Received 08 Jan 2017, Accepted 24 Mar 2017, Published online: 05 Oct 2017

ABSTRACT

Traumatic brain injury (TBI) is a widely recognized risk factor for neurodegenerative disease. The purpose of this review is to provide an update on the state of the science related to injury cascades in TBI-related neurodegeneration. Acute and chronic pathological outcomes of TBI are similar to those seen in several neurodegenerative conditions, suggesting common linking pathways. Initial research described severe TBI patients with post-mortem identification of abnormal proteins, such as amyloid deposits. History of mild TBI (mTBI) is less consistently associated with heightened risk of neurodegenerative outcomes, but specific populations with complicated medical histories and comorbidities may be more susceptible. Our understanding of a pathological signature associated with repetitive mTBI and/or subclinical brain trauma advanced significantly over the past decade, and is now commonly referred to as chronic traumatic encephalopathy. We discuss hypotheses linking TBI to neurodegenerative disease, and the importance of considering factors like injury severity, timing of injury (early life versus older age), injury frequency, and repetitive subclinical brain trauma when extrapolating results from current literature to certain populations. We describe the challenges to obtaining the data necessary for accurate epidemiological research and determination of true risk magnitude, and note the importance of developing treatment-based approaches to risk mitigation.

Introduction

Traumatic brain injury (TBI) and neurodegenerative disease are both international public health concerns. Heightened research efforts investigating links between these two conditions indicate TBI may elevate risk of developing neurodegenerative pathology by a variety of potential mechanisms. Harrison Martland first described poor long-term outcome specific to repetitive TBI mechanisms as ‘punch drunk’ syndrome in 1928, later termed dementia pugilistica by Millspaugh in 1937 and ‘chronic progressive traumatic encephalopathy of boxers’ by Critchley in 1957 [1−3]. Chronic traumatic encephalopathy (CTE) is now a widely recognized term among the public that was popularized over the past decade because of evidence suggesting athletes and military personnel exposed to repetitive brain trauma are at risk of the neurodegenerative pathology [Citation4,Citation5]. Studies also identify TBI as a possible environmental risk factor for more common degenerative diseases such as Alzheimer’s disease (AD). Overlapping neuropathological outcomes between TBI and neurodegenerative disorders lend support to this association, though a great deal remains unknown. Here, we note population and injury-severity-specific considerations, and we briefly review hypothesized mechanisms, epidemiological studies describing diagnosed neurodegenerative disease and dementia after TBI exposure, and future research avenues.

TBI severity and at-risk populations

TBI is described along a continuum from mild to moderate to severe based on gross neurological assessment and neuroimaging findings. Many consider ‘concussion’ synonymous with mild TBI (mTBI). Recent concerns have emerged about repetitive subclinical brain trauma (RSBT), or forces transmitted to the brain that do not cross a threshold of observable or measurable functional changes but still may result in shear-strain of axons. These concerns are likely due to findings that repetitive trauma in certain sports (American football, ice hockey and others) and soldiers has manifested as CTE or structural brain changes that do not necessarily correlate with a significant, documented concussion history. Civilians are at risk of single-event TBI of all severities via mechanisms such as motor vehicle accidents, assault, or falls, while certain collision sport athletes and military personnel are uniquely exposed to RSBT as well as more frequent mTBI. The degree to which findings from acute, severe TBI patients directly apply to those with extensive RSBT or multiple mTBI, and vice versa is unclear, but such data likely identify candidate mechanisms underlying the relationships to later-life neurodegenerative disease.

Acute pathophysiological effects of single-event TBI

TBI results in diffuse axonal injury characterized by mechanical deformation of axons and subsequent indiscriminate glutamate release, spreading depolarization, and altered neuronal metabolism [Citation6]. Dysregulated axonal transport systems lead to axonal swelling that may result in deprivation of necessary proteins at axon terminals and, ultimately, focal nerve disconnection (i.e. ‘secondary axotomy’) or reduced elasticity [Citation7]. Excess amount of proteins associated with TBI, beta-amyloid (Aβ) and phosphorylated tau (pTau), are implicated in many neurodegenerative diseases. Johnson et al. [Citation8] and Washington et al. [Citation9] review axonal and cellular polypathologies following TBI in greater detail.

Human TBI studies indicate that damaged axons ‘can serve as a large reservoir’ of amyloid precursor protein (APP) and Aβ [Citation10]. APP levels in the temporal cortex increase within hours after severe TBI in humans and localize to axonal swellings, neurites, and cell bodies in the absence of acute neurofibrillar pathology [Citation11]. APP upregulation and accumulation is an established marker of diffuse axonal injury in TBI, significantly increasing risk for neurotoxic levels of Aβ [Citation12,Citation13]. Roberts and colleagues indicated approximately 30% of severe TBI patients test positive for Aβ plaques, and a significant effect of age such that incidence was 20% in those younger than age 50 and 60% in patients between 51 and 60 [Citation14]. Studies also show significantly higher incidence of intracellular and intra-axonal accumulation of non-plaque Aβ [Citation11,Citation15]. Similar results are noted in animal studies using fluid percussion and controlled cortical impact injury models [Citation16Citation21].

The shear-strain force from TBI may result in hyperphosphorylation, misfolding, and accumulation of tau, which disrupts axonal transport systems. The presence of aggregated pTau as neurofibrillary tangles (NFTs) acutely after a single TBI is not as common as Aβ, but the distribution of tau pathology appears unique to TBI aetiology. Uryu et al. [Citation22] examined brain tissue from 18 cases following single incident fatal TBI and found far less prevalence of pTau relative to APP accumulation within axons. A more recent study compared NFT deposition in patients diagnosed with only a single TBI (with 1–47 years’ survival) and found no difference in prevalence compared to an age-matched control group. However, the TBI group displayed NFTs more frequently when limiting analyses to subjects 60 years or younger. Deposition of age-related NFTs or tau deposition does not occur commonly in subjects below 60 years. The TBI group also had a unique distribution of NFT depositions in the cortical layers – clustering at depths of cortical sulci, compared to controls exhibiting NFTs predominantly in entorhinal cortex and the hippocampus, considered mostly consistent with normal aging [Citation23]. Cortical NFTs are not widely seen with normal ageing, but with clinically manifest AD.

Hypotheses linking tbi to progressive neurodegenerative Disease

Mechanisms driving the potential neurodegenerative effects of acute TBI, multiple mTBI, or RSBT are poorly understood. Proposed hypotheses include decreased cognitive reserve, chronic inflammation, chronic microglia activation, acute upregulation of APP and subsequent AD-like cascades, and slow degeneration of axonal connections due to altered protein degradation processes. The widespread effects of TBI on neuronal homeostasis and regulatory functions suggest one or many of these hypotheses may drive chronic dysfunction even in the absence of subsequent or repetitive forces transmitted to the brain.

The cognitive reserve hypothesis holds that the effects of TBI modify the ‘normal ageing’ trajectory for the affected individual, and multiple injuries may have synergistic negative effects that amplify or accelerate the risk of crossing a threshold for dementia diagnosis. Neuropathologically, acute neuroinflammation after TBI is associated with cytokine release and persistent microglia activation, evidenced by reactive microglia found post-mortem months to years after a single TBI [Citation24Citation27]. Johnson and colleagues found reactive microglia present in 28% of brains examined over a year following a single TBI, which supports previous data indicating chronic inflammation and microglia activation up to 17 years after TBI in areas distal to the trauma locus [Citation25,Citation28].

As previously mentioned, acute TBI pathology can include Aβ and tau deposition. The normally functioning brain has mechanisms for protein degradation and removal. The ubiquitin-proteasome pathway, for example, is an intracellular mechanism that regulates degradation of both normal and abnormal proteins, and promotes normal cell growth and metabolism. This pathway may be impaired due to chronic inflammation following TBI, resulting in inability to clear proteins such as Aβ and pTau efficiently [Citation29]. The combination of abnormal protein deposition and reduced degradation and clearance abilities suggests plausible mechanisms linking single-event moderate to severe TBI to neurodegenerative processes.

Repetitive brain trauma and neurodegeneration

Athletes participating in high-frequency collision sports and certain military personnel are exposed to RSBT and potentially multiple mTBI [Citation30]. CTE is the primary neurodegenerative tauopathy associated with RSBT and multiple mTBI history, though other proteomic molecules such as Aβ and TAR DNA-binding protein 43 (TDP-43) are also frequently observed. Recently published pathological criteria defined pTau aggregates in neurons, astrocytes, and cell processes around small vessels in cluster patterns at the depths of cortical sulci as the pathognomonic sign of CTE [Citation31]. Continuous tau deposition in the cortical lamina differentiated AD from CTE, as does the absence, in CTE, of NFT deposition in other parts of the cortex and limbic system, particularly in milder cases of CTE. However, data show that up to 37% of individuals diagnosed with CTE pathology also met criteria for comorbid neurodegenerative pathology including AD, Lewy Body Disease, and frontotemporal lobar degeneration [Citation5], largely in older subjects, and CTE has been identified almost exclusively in individuals with a history of repetitive brain trauma, though some recent findings raise questions of whether such exposure is necessary [Citation32,Citation33]. Lack of prospective longitudinal studies or validated in vivo diagnostic markers, and current sample biases significantly limit accurate determination of CTE incidence and prevalence. Literature to date has not described CTE as an outcome of single-event TBI.

The absence of premortem CTE diagnostic markers complicates attribution of structural and functional changes seen in individuals with exposure to repetitive brain trauma, and thus, definitive diagnosis. Coughlin and colleagues found prolonged neuroinflammation in former National Football League (NFL) players associated with hippocampal atrophy as well as evidence of activated microglia years after retirement from sport [Citation34], like previously described outcomes after single-event TBI. Although early case studies using positron emission tomography (PET) revealed uptake of tau-binding ligands in retired NFL players with retention distribution inconsistently characteristic of expected CTE patterns [Citation35,Citation36], more recent reports do show a pattern consistent with the pathological distribution associated with CTE. Increased cortical thinning, enlarged ventricles, subcortical atrophy, and diffuse white matter abnormalities have all been reported in retired NFL athletes [Citation37Citation39]. Blood and cerebrospinal fluid (CSF) markers of central nervous system (CNS) injury may also provide insight to pathophysiological outcomes of RSBT and multiple mTBI. A recent study found a higher number of tau-positive exosomes in former American football athletes compared to retired non-collision sport athletes [Citation40]. Alosco et al. [Citation41] reported a weak correlation between head impact exposure and plasma total tau levels, but no relationship with clinical outcomes. Other studies demonstrated evidence of several markers of acute axonal injury following repetitive impacts sustained during a boxing match, but no differences between boxers and controls after 3 months of rest except for neurofilament light protein levels [Citation42].

Risk of dementia and neurodegeneration following TBI

Interpreting studies describing risk of dementia or neurodegenerative disease following TBI requires attention to time since injury (e.g. remote TBI history versus TBI in older adults), injury severity, and single-event versus repetitive injury (multiple mTBI and/or RSBT). There appears to be a weak statistical association between remote mTBI with loss of consciousness and incident dementia, on the order of 1.5 to 2.0-fold increased risk [Citation43,Citation44], with some evidence suggesting mTBI is a weaker predictor than many other neurobiological and psychosocial risk factors [Citation45]. Other studies, however, have found no association [Citation46,Citation47]. Crane and colleagues found no association between remote mTBI and dementia or AD pathology, but reported a specific heightened risk of Lewy body pathology [Citation48]. Mayeux et al. [Citation49] also found no association between TBI history and AD diagnosis, but reported a twofold higher risk in individuals with apolipoprotein epsilon-4 (ApoE-ε4) and a tenfold increased risk in those with both history of TBI and ApoE-ε4, raising the possibility of synergistic effects of TBI and genetic predisposition for neurodegenerative disease [Citation50].

Data are similar in studies of older adults sustaining a TBI, with reported elevated risks ranging from 1.3 to 3.0 times greater likelihood of subsequent dementia diagnosis [Citation51,Citation52] and even higher risk in older individuals with the ApoE-ε4 genotype [Citation53]. Gardner and colleagues assessed risk for dementia following TBI in older adults (minimum age 55) compared to older adults sustaining an orthopaedic injury without brain trauma. They found that moderate to severe TBI was associated with increased dementia risk across all age groups, and mTBI was a significant predictor in older subjects only (65 and older) [Citation54]. Thus, consideration of injury age, severity, and genetic interactions may be important for determining risk of neurodegenerative disease following TBI.

Studies of repetitive brain trauma associated with collision sport participation have shown inconsistent risks of later-life neurodegenerative disease or dementia. Lehman et al. [Citation55] found retired NFL players were at lower overall risk of mortality compared to age-matched controls, but had a threefold increased risk of having either AD or amyotrophic lateral sclerosis (ALS) at death. Two recent Mayo Clinic studies found no association between high school football participation and subsequent risk of dementia, Parkinson’s disease (PD), or ALS, suggesting that risk was related to the degree of brain trauma exposure and length of playing career [Citation56,Citation57]. Like outcomes reported in single-event TBI studies, Jordan and colleagues described an interaction of genetics and brain trauma exposure such that the ApoE-ε4 genotype was associated with worse outcome in boxers with a high amount of exposure [Citation58]. Maroon et al. [Citation59] systematically reviewed the initial 153 case reports of CTE and found no relationship between CTE and age of death or ApoE allele type, and concluded that significant questions remain regarding the popularized belief of ‘widespread existence of CTE in contact sports’. Incidence, prevalence, and risk factors for CTE are currently unknown due to the absence of longitudinal studies of representative populations; the clear majority of brains analysed from subjects suffering multiple recurrent TBI were symptomatic before death and came to autopsy because of their clinical symptoms.

Challenges and future directions

Getting precise, quantitative head impact data from football and other collision sports, as well as military combat settings, is difficult. Mechanistic differences between direct impacts seen in collision sports versus blast wave forces, and blast wave forces plus collision injury, in combat must also be studied. For example, a recent report from Shively and colleagues described potentially distinct pathological signatures associated with chronic blast exposure – specifically, astroglial scarring in the subpial glial plate, cortical blood vessels, grey-white matter junctions, and periventricular regions [Citation60]. These findings were absent in cases of individuals with exposure to a single-event TBI, though a previous investigation from Goldstein et al. [Citation61] reported evidence of widespread astrocytosis in mice exposed to a single blast injury.

Co-occurring posttraumatic stress disorder (PTSD) is frequently a confounding issue in military settings, and retired collision sport athletes often have comorbid neurobiological and psychosocial conditions complicating the appraisal and attribution of cognitive, mood, and behavioural difficulties experienced after retirement [Citation62]. Future research efforts must utilize prospective, longitudinal, and epidemiological designs for determining incidence and prevalence of CTE and other neurodegenerative diseases in populations at risk for TBI and/or RSBT relative to the general population. Advanced neuroimaging (e.g. PET [Citation63]) and physiological biomarkers (e.g. CSF, serum, and plasma concentrations of CNS injury-related proteins) coupled with comprehensive neuropsychological evaluations will assist in defining clinical signatures of CTE and possible phenotypic subtypes. Family-based and case-control genetic studies may elucidate gene-environment interactions and identify individuals at higher risk of adverse outcome following TBI both acutely and chronically.

Lastly, and perhaps most importantly, treatment modalities must be studied targeting not only aggregation and propagation of neurodegenerative proteins such as tau and Aβ [Citation16,Citation64], but also modifiable risk factors believed to contribute to disease progression and overall quality of life, such as cardiovascular health, sleep disorders, psychiatric difficulties, and substance use, among others [Citation62]. Models of the time course of AD pathology indicate that amyloid alteration precedes tau abnormalities by years, and that ongoing, chronic deposition processes may be occurring for a decade or more before observed evidence of cognitive or functional changes. Thus, it may be important to consider TBI as a risk factor for neuropathological outcomes separately from clinical outcomes (i.e. dementia), while also recognizing the role of prophylactic interventions that may prevent, delay, or slow formation of pathologic protein aggregates; and slow progression of disease through preservation or enhancement of neural structure and cognitive reserve.

Declaration of interest

This work was supported by NIA AG047266 (STDeK). Dr DeKosky reports consulting for Acumen, Inc., Amgen, Biogen, Cognition Therapeutics, Lilly Pharmaceuticals and Novartis Pharmaceuticals.

References

  • Millspaugh J. Dementia pugilistica. US Naval Med Bull. 1937;35:297–303.
  • Critchley M. Medical aspects of boxing, particularly from a neurological standpoint. Br Med J. 1957;1(5015):357.
  • Martland HS. Punch drunk. J Am Med Assoc. 1928;91(15):1103–1107.
  • Omalu BI, DeKosky ST, Minster RL, Kamboh MI, Hamilton RL, Wecht CH. Chronic traumatic encephalopathy in a National Football League player. Neurosurgery. 2005;57(1):128–134; discussion 128–134.
  • McKee AC, Stein TD, Nowinski CJ, Stern RA, Daneshvar DH, Alvarez VE, Lee HS, Hall G, Wojtowicz SM, Baugh CM, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain. 2013;136(1):43–64.
  • Giza CC, Hovda DA. The new neurometabolic cascade of concussion. Neurosurgery. 2014;75 Suppl 4:S24–33.
  • Christman CW, Grady MS, Walker SA, Holloway KL, Povlishock JT. Ultrastructural studies of diffuse axonal injury in humans. J Neurotrauma. 1994;11(2):173–186.
  • Johnson VE, Stewart W, Smith DH. Axonal pathology in traumatic brain injury. Exp Neurol. 2013;246:35–43.
  • Washington PM, Villapol S, Burns MP. Polypathology and dementia after brain trauma: Does brain injury trigger distinct neurodegenerative diseases, or should they be classified together as traumatic encephalopathy? Exp Neurol. 2016;275:381–388.
  • Smith DH, Chen X-h, Iwata A, Graham DI. Amyloid β accumulation in axons after traumatic brain injury in humans. J Neurosurg. 2003;98(5):1072–1077.
  • Ikonomovic MD, Uryu K, Abrahamson EE, Ciallella JR, Trojanowski JQ, Lee VM, Clark RS, Marion DW, Wisniewski SR, DeKosky, ST. Alzheimer’s pathology in human temporal cortex surgically excised after severe brain injury. Exp Neurol. 2004;190(1):192–203.
  • DeKosky ST, Abrahamson EE, Ciallella JR, Paljug WR, Wisniewski SR, Clark RS, Ikonomovic MD. Association of increased cortical soluble Aβ42 levels with diffuse plaques after severe brain injury in humans. Arch Neurol. 2007;64(4):541–544.
  • Mielke MM, Savica R, Wiste HJ, Weigand SD, Vemuri P, Knopman DS, Lowe VJ, Roberts RO, Machulda MM, Geda YE, et al. Head trauma and in vivo measures of amyloid and neurodegeneration in a population-based study. Neurology. 2014;82(1):70–76.
  • Roberts G, Gentleman S, Lynch A, Murray L, Landon M, Graham D. Beta amyloid protein deposition in the brain after severe head injury: implications for the pathogenesis of Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1994;57(4):419–425.
  • Chen XH, Johnson VE, Uryu K, Trojanowski JQ, Smith DH. A lack of amyloid β plaques despite persistent accumulation of amyloid β in axons of long‐term survivors of traumatic brain injury. Brain Pathol. 2009;19(2):214–223.
  • Loane DJ, Pocivavsek A, Moussa CE, Thompson R, Matsuoka Y, Faden AI, Rebeck GW, Burns MP. Amyloid precursor protein secretases as therapeutic targets for traumatic brain injury. Nat Med. 2009;15(4):377.
  • Masumura M, Hata R, Uramoto H, Murayama N, Ohno T, Sawada T. Altered expression of amyloid precursor proteins after traumatic brain injury in rats: in situ hybridization and immunohistochemical study. J Neurotrauma. 2000;17(2):123–134.
  • Murakami N, Yamaki T, Iwamoto Y, Sakakibara T, Kobori N, Fushiki S, Ueda S. Experimental brain injury induces expression of amyloid precursor protein, which may be related to neuronal loss in the hippocampus. J Neurotrauma. 1998;15(11):993–1003.
  • Pierce J, Trojanowski JQ, Graham DI, Smith DH, McIntosh TK. Immunohistochemical characterization of alterations in the distribution of amyloid precursor proteins and beta-amyloid peptide after experimental brain injury in the rat. J Neurosci. 1996;16(3):1083–1090.
  • Washington PM, Morffy N, Parsadanian M, Zapple DN, Burns MP. Experimental traumatic brain injury induces rapid aggregation and oligomerization of amyloid-beta in an Alzheimer’s disease mouse model. J Neurotrauma. 2014;31(1):125–134.
  • Tran HT, LaFerla FM, Holtzman DM, Brody DL. Controlled cortical impact traumatic brain injury in 3xTg-AD mice causes acute intra-axonal amyloid-β accumulation and independently accelerates the development of tau abnormalities. J Neurosci. 2011;31(26):9513–9525.
  • Uryu K, Chen X-H, Martinez D, Browne KD, Johnson VE, Graham DI, Lee VM, Trojanowski JQ, Smith DH. Multiple proteins implicated in neurodegenerative diseases accumulate in axons after brain trauma in humans. Exp Neurol. 2007;208(2):185–192.
  • Johnson VE, Stewart W, Smith DH. Widespread tau and amyloid‐beta pathology many years after a single traumatic brain injury in humans. Brain Pathol. 2012;22(2):142–149.
  • Ziebell JM, Morganti-Kossmann MC. Involvement of pro-and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics. 2010;7(1):22–30.
  • Johnson VE, Stewart JE, Begbie FD, Trojanowski JQ, Smith DH, Stewart W. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain. 2013;136(1):28–42.
  • Gentleman S, Leclercq P, Moyes L, Graham DI, Smith C, Griffin WS, Nicoll JA. Long-term intracerebral inflammatory response after traumatic brain injury. Forensic Sci Int. 2004;146(2):97–104.
  • Smith C, Gentleman SM, Leclercq PD, Murray LS, Griffin WS, Graham DI, Nicoll JA. The neuroinflammatory response in humans after traumatic brain injury. Neuropathol Appl Neurobiol. 2013;39(6):654–666.
  • Ramlackhansingh AF, Brooks DJ, Greenwood RJ, Bose SK, Turkheimer FE, Kinnunen KM, Gentleman S, Heckermann RA, Gunanayagam K, Gelosa G, et al. Inflammation after trauma: microglial activation and traumatic brain injury. Ann Neurol. 2011;70(3):374–383.
  • Figueiredo-Pereira ME, Rockwell P, Schmidt-Glenewinkel T, Serrano P. Neuroinflammation and J2 prostaglandins: linking impairment of the ubiquitin-proteasome pathway and mitochondria to neurodegeneration. Front Mol Neurosci. 2015;7:104.
  • DeKosky ST, Ikonomovic MD, Gandy S. Traumatic brain injury–football, warfare, and long-term effects. N Engl J Med. 2010;363(14):1293–1296.
  • McKee AC, Cairns NJ, Dickson DW, Folkerth RD, Keene CD, Litvan I, Perl DP, Stein TD, Vonsattel JP, Stewart W, et al. The first NINDS/NIBIB consensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol. 2016;131(1):75–86.
  • Koga S, Dickson DW, Bieniek KF. Chronic traumatic encephalopathy pathology in multiple system atrophy. J Neuropathol Exp Neurol. 2016;75(10):963–970.
  • Noy S, Krawitz S, Del Bigio MR. Chronic traumatic encephalopathy-like abnormalities in a routine neuropathology service. J Neuropathol Exp Neurol. 2016;75(12):1145–1154.
  • Coughlin JM, Wang Y, Munro CA, Ma S, Yue C, Chen S, Airan R, Kim PK, Adams AV, Garcia C, et al. Neuroinflammation and brain atrophy in former NFL players: An in vivo multimodal imaging pilot study. Neurobiol Dis. 2015;74:58–65.
  • Dickstein D, Pullman M, Fernandez C, Short J, Kostakoglu L, Knesaurek K, Jordan B, Gordon W, Dams-O’Connor K, Delman B, et al. Structural MRI and molecular PET imaging (AV45 and AV1451) in the diagnosis of chronic traumatic encephalopathy in vivo: Study of a retired NFL player (P4. 046). Neurology. 2016;86( 16Supplement): P4.046.
  • Mitsis E, Riggio S, Kostakoglu L, Dickstein DL, Machac J, Delman B, Goldstein M, Jennings D, D’Antonio E, Martin J, et al. Tauopathy PET and amyloid PET in the diagnosis of chronic traumatic encephalopathies: studies of a retired NFL player and of a man with FTD and a severe head injury. Transl Psychiatry. 2014;4(9):e441.
  • Tremblay S, De Beaumont L, Henry LC, Boulanger Y, Evans AC, Bourgouin P, Poirier J, Theoret H, Lassonde M. Sports concussions and aging: a neuroimaging investigation. Cerebral Cortex. 2013;23(5):1159–1166.
  • Tremblay S, Henry LC, Bedetti C, Larson-Dupuis C, Gagnon JF, Evans AC, Theoret H, Lassonde M, De Beaumont L. Diffuse white matter tract abnormalities in clinically normal ageing retired athletes with a history of sports-related concussions. Brain. 2014;137(Pt 11):2997–3011.
  • Strain JF, Womack KB, Didehbani N, Spence JS, Conover H, Hart J, Kraut MA, Cullum CM. Imaging correlates of memory and concussion history in retired national football league athletes. JAMA Neurol. 2015;72(7):773–780.
  • Stern RA, Tripodis Y, Baugh CM, Fritts NG, Martin BM, Chaisson C, Cantu RC, Joyce JA, Shah S, Ikezu T, et al. Preliminary study of plasma exosomal tau as a potential biomarker for chronic traumatic encephalopathy. J Alzheimer’s Dis. 2016;51(4):1099–1109.
  • Alosco ML, Tripodis Y, Jarnagin J, Baugh CM, Martin B, Chaisson CE, Estochen N, Song L, Cantu RC, Jeromin A, et al. Repetitive head impact exposure and later-life plasma total tau in former NFL players. Alzheimer’s Dementia: Diagn Assess Dis Monit. 2017;7:33–40.
  • Zetterberg H, Hietala MA, Jonsson M, Andreasen N, Styrud E, Karlsson I, Edman A, Popa C, Rasulzada A, Wahlund LO, et al. Neurochemical aftermath of amateur boxing. Arch Neurol. 2006;63(9):1277–1280.
  • Mortimer J, Van Duijn C, Chandra V, Fratiglioni L, Graves AB, Heyman A, Jorm AF, Kokmen E, Kondo K, Rocca WA, et al. Head trauma as a risk factor for Alzheimer’s disease: a collaborative re-analysis of case-control studies. Int J Epidemiol. 1991;20(Supplement 2):S28–S35.
  • Fleminger S, Oliver D, Lovestone S, Rabe-Hesketh S, Giora A. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. J Neurol Neurosurg Psychiatry. 2003;74(7):857–862.
  • Nordström P, Michaëlsson K, Gustafson Y, Nordström A. Traumatic brain injury and young onset dementia: a nationwide cohort study. Ann Neurol. 2014;75(3):374–381.
  • Plassman BL, Havlik RJ, Steffens DC, Helms MJ, Newman TN, Drosdick D, Phillips C, Gau BA, Welsh-Bohmer KA, Burke JR, et al. Documented head injury in early adulthood and risk of Alzheimer’s disease and other dementias. Neurology. 2000;55(8):1158–1166.
  • Dams-O’Connor K, Gibbons LE, Bowen JD, McCurry SM, Larson EB, Crane PK. Risk for late-life re-injury, dementia and death among individuals with traumatic brain injury: a population-based study. J Neurol Neurosurg Psychiatry. 2013;84(2):177–182.
  • Crane PK, Gibbons LE, Dams-O’Connor K, Trittschuh E, Leverenz JB, Keene CD, Sonnen J, Montine TJ, Bennet DA, Leurgans S, et al. Association of traumatic brain injury with late-life neurodegenerative conditions and neuropathologic findings. JAMA Neurol. 2016;73(9):1062–1069.
  • Mayeux R, Ottman R, Maestre G, Ngai C, Tang MX, Ginsberg H, Chun M, Tycko B, Shelanski M. Synergistic effects of traumatic head injury and apolipoprotein-epsilon4 in patients with Alzheimer’s disease. Neurology. 1995;45(3):555–557.
  • Gandy S, DeKosky ST. APOE ε4 status and traumatic brain injury on the gridiron or the battlefield. Sci Transl Med. 2012;4(134):134ed134–134ed134.
  • Perry DC, Sturm VE, Peterson MJ, Pieper CF, Bullock T, Boeve BF, Miller BL, Guskiewicz KM, Berger MS, Kramer JH. Association of traumatic brain injury with subsequent neurological and psychiatric disease: a meta-analysis. J Neurosurg. 2016;124(2):511–526.
  • Lee Y-K, Hou S-W, Lee C-C, Hsu C-Y, Huang Y-S, Su Y-C. Increased risk of dementia in patients with mild traumatic brain injury: a nationwide cohort study. PloS One. 2013;8(5):e62422.
  • Luukinen H, Viramo P, Herala M, Kervinen K, Kesaniemi YA, Savola O, Winqvist S, Jokelainen J, Hillborn M. Fall‐related brain injuries and the risk of dementia in elderly people: a population‐based study. Eur J Neurol. 2005;12(2):86–92.
  • Gardner RC, Burke JF, Nettiksimmons J, Kaup A, Barnes DE, Yaffe K. Dementia risk after traumatic brain injury vs nonbrain trauma: the role of age and severity. JAMA Neurol. 2014;71(12):1490–1497.
  • Lehman EJ, Hein MJ, Baron SL, Gersic CM. Neurodegenerative causes of death among retired National Football League players. Neurology. 2012;79(19):1970–1974.
  • Savica R, Parisi JE, Wold LE, Josephs KA, Ahlskog JE. High school football and risk of neurodegeneration: a community-based study. Mayo Clinic Proceedings. 2012;87(4):335–340.
  • Janssen PH, Mandrekar J, Mielke MM, Ahlskog JE, Boeve BF, Josephs K, Savica R. High School Football and Late-Life Risk of Neurodegenerative Syndromes 1956–1970., Mayo Clinic Proceedings. 2017;92(1):66–71.
  • Jordan BD, Relkin NR, Ravdin LD, Jacobs AR, Bennett A, Gandy S. Apolipoprotein E epsilon4 associated with chronic traumatic brain injury in boxing. JAMA. 1997;278(2):136–140.
  • Maroon JC, Winkelman R, Bost J, Amos A, Mathyssek C, Miele V. Chronic traumatic encephalopathy in contact sports: a systematic review of all reported pathological cases. PloS One. 2015;10(2):e0117338.
  • Shively SB, Horkayne-Szakaly I, Jones RV, Kelly JP, Armstrong RC, Perl DP. Characterisation of interface astroglial scarring in the human brain after blast exposure: a post-mortem case series. Lancet Neurol. 2016;15(9):944–953.
  • Goldstein LE, Fisher AM, Tagge CA, Zhang XL, Velisek L, Sullivan JA, Upreti C, Kracht JM, Ericsson M, Wojnarowicz MW, Goletiani CJ. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci Transl Med. 2012;4(134):134 ra60-.
  • Asken BM, Sullan MJ, Snyder AR, Houck ZM, Bryant VE, Hizel LP, McLaren ME, Dede DE, Jaffee MS, DeKosky ST, et al. Factors influencing clinical correlates of chronic traumatic encephalopathy (CTE): a review. Neuropsychol Rev. 2016;26(4):340–363.
  • Gandy S, DeKosky ST. [18F]-T807 tauopathy PET imaging in chronic traumatic encephalopathy. F1000Research. 2014;3229. doi:10.12688/f1000research.5372.1. eCollection 2014.
  • Abrahamson EE, Ikonomovic MD, Dixon CE, DeKosky ST. Simvastatin therapy prevents brain trauma‐induced increases in β‐amyloid peptide levels. Ann Neurol. 2009;66(3):407–414.