Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Journal of Clinical Neuroscience 41 (2017) 24–29

Contents lists available at ScienceDirect

Journal of Clinical Neuroscience


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

Review article

Chronic Traumatic Encephalopathy: The cellular sequela to repetitive


brain injury
Alexander R. Vile a,⇑, Leigh Atkinson b
a
James Cook University College of Medicine and Dentistry, Australia
b
Wesley Pain and Spine Centre, Brisbane, QLD, Australia

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

Article history: This review aims to integrate current literature on the pathogenic mechanisms of Chronic Traumatic
Received 12 January 2017 Encephalopathy (CTE) to create a multifactorial understanding of the disease. CTE is a progressive neu-
Accepted 6 March 2017 rodegenerative disease, classed as a tauopathy, although it appears the pathogenic mechanisms are more
complex than this. It affects those with a history of repetitive mild traumatic brain injury. Currently, there
are no treatments for CTE and the disease can only be affirmatively diagnosed in post mortem.
Keywords: Understanding the pathogenesis of the disease will provide an avenue to explore possible treatment
Chronic Traumatic Encephalopathy
and diagnostic modalities. The pathological hallmarks of CTE have been well characterised and have been
CTE
Tau
linked to the pathophysiologic mechanisms in this review. Human studies are limited due to ethical
Immune excitotoxicity implications of exposing subjects to head trauma. Phosphorylation of tau, microglial activation, TAR
Mild traumatic brain injury DNA-binding protein 43 and diffuse axonal injury have all been implicated in the pathogenesis of CTE.
TAR DNA-binding protein 43 The neuronal loss and axonal dysfunction mediated by these pathognomonic mechanisms lead to the
Diffuse axonal injury broad psycho-cognitive symptoms seen in CTE.
Ó 2017 Elsevier Ltd. All rights reserved.

1. Introduction This is accompanied by marked atrophy of the temporal and fron-


tal lobes, diencephalon and brainstem, with depigmentation of the
Chronic Traumatic Encephalopathy (CTE) is a progressive neu- locus coeruleus and substantia nigra [2,5–7]. Axonal pathology in
rodegenerative disease, pathologically marked by tauopathy of the early stages is defined by distorted axonal varicosities, pro-
the brain. The disease was first described by Martland (1928), gressing to axonal loss in the cortex, subcortical white matter,
who introduced the term ‘‘punch drunk” to medical literature to and deep white matter tracts of the diencephalon in later stages
describe a collection of symptoms, including Parkinsonism, mental [2]. Tar-DNA-binding protein (TDP-43) pathology is seen through-
confusion and psychiatric illness among retired boxers [1]. Epi- out all stages, though not in all patients, but is universal by stage
demiological evidence has established repetitive head trauma as IV, with 10% of patients also developing a motor neuron disease
the aetiology of the disease, with the disease most prominent in indistinguishable from Amyotrophic Lateral Sclerosis [2]. Neuronal
boxers and American football players [2,3]. Based on a 2013 case and axonal loss leads to the symptomatology, shown in Table 1.
series of 68 patients, the largest to date, McKee et.al. established Despite having characterised the pathological hallmarks of the dis-
a pathological grading system of I–IV, with this correlating with ease, the pathophysiological events leading to these hallmarks
the severity of symptoms. To summarise macroscopically, milder remain poorly understood. This review aims to integrate current
cases are marked by cavum septum pellucidum and mild enlarge- understanding of the multiple mechanisms involved in the patho-
ment of the frontal and temporal horns of the lateral ventricles, genesis of CTE to create a multifactorial model of understanding
accompanied by microscopic neurofibrillary tangles located the disease.
perivascular and at the depths of cerebral sulci [2]. These two
microscopic findings are what distinguish CTE from other tauopa- 2. Pathogenesis of CTE
thies [2,4]. Neurofibrillary tangles then spread from the depth of
the sulci and perivascular to the adjacent superficial cortical layers. Much of the information regarding the pathogenesis of CTE has
been derived from animal studies. Whilst the pathological hall-
⇑ Corresponding author at: 8 Rivergum Place, Fig Tree Pocket, Queensland 4069, marks of the disease have been well characterized in humans,
Australia. the pathogenesis has been defined to a lesser extent due to the eth-
E-mail address: vile.alex07@gmail.com (A.R. Vile). ical implications of exposing human subjects to head trauma. It is

http://dx.doi.org/10.1016/j.jocn.2017.03.035
0967-5868/Ó 2017 Elsevier Ltd. All rights reserved.
A.R. Vile, L. Atkinson / Journal of Clinical Neuroscience 41 (2017) 24–29 25

Table 1 notion that hyperphosphorylated tau causes neuronal death, as


Symptomatology of CTE [8]. indicated by genetic studies implicating tau as the sole cause of
Cognitive features Behavioural Mood Motor neuronal death in Fronto-temporal dementia and parkinsonism
features features features linked to chromosome 17, another tauopathy [24]. Based on the lit-
Memory impairment Physical violence Depression Ataxia erature, the argument presented in this review is that tau oligo-
Executive Verbal violence Hopelessness Dysarthria mers are the more toxic of the two, yet NFTs represent an
dysfunction adaptive response to prevent the development of toxic oligomers,
Impaired attention Explosivity Suicidality Parkinsonism
Dysgraphia Loss of control Anxiety Gait
but ultimately lead to neurodegeneration [22,25]. The aforemen-
Lack of insight Short fuse Fearfulness Tremor tioned has been difficult to study given tau does not readily aggre-
Preservation Impulsivity Irritability Masked gate into filaments within an ideal time frame for culture studies or
facies within an animal’s short lifespan, though it is thought that the
Language difficulties Paranoid delusions Apathy Rigidity
hyperphosphorylation of tau leads to a toxic gain of function with
Dementia Aggression Loss of Weakness
interest a loss of function, with this accounting for neuronal death. How-
Alogia Rage Labile Spasticity ever, it is not certain whether this toxicity is exerted by NFTs or oli-
emotions gomers [22,25]. Case control studies from the 1970s and 1980s,
Visuospatial Inappropriate Fatigue Clonus correlated the severity of cognitive impairments in humans to
difficulties speech
the extent of NFT formation, which intuitively indicated its role
Cognitive Boastfulness Flat affect
impairment in neurodegeneration [26–28]. This older paradigm has been chal-
Reduced intelligence Childish behaviour Insomnia lenged in a review by Crespo-Biel (2012), which show memory def-
Socially Mania icits preceding NFT pathology, indicating a toxic intermediate in
inappropriate
the tau phosphorylation cascade is likely to blame for neuronal
Disinhibited Euphoria
behaviour damage [29].
Psychosis Mood swings Studies into the mechanism by which these tau oligomers cause
Social isolation Prolix cells death are limited. One study has implicated tau oligomers in
interfering with anterograde axonal transport system protein kine-
sin, leading to an inability to maintain axons and axonal degener-
not yet clear the degree to which these studies are applicable on ation [30]. This would explain the milder cognitive impairments
humans, but they do provide a model for the disease. Phosphory- seen early in the disease, without the presence of pathological hall-
lated tau deposition, TAR DNA-binding protein 43 (TDP-43), micro- marks. Neuronal loss can be explained by the resultant deaf-
glial activation and diffuse axonal injury have been implicated in ferentation leading to a lack of survival signalling, known as
the pathogenesis of CTE. anterograde transneural degeneration [31]. Some studies have
addressed the role of caspase activation in tauopathy, however
the findings of these studies are inconsistent. Active caspases have
2.1. Tau phosphorylation been identified using fluorescent antibody tagging in neurons con-
taining NFT’s and shown to cause apoptosis in cultured hippocam-
Phosphorylation of tau has been suggested to play a major role pal neurons [32–34]. However, other studies have argued against
in the pathogenesis of CTE. The notion that tau has been implicated this theory, suggesting apoptosis was not an important contribut-
as a pathogenic mechanism has arisen in rat studies, in which ing factor to neuronal death, rather caspases cleaving tau increases
injection of oligomeric tau into the brains on healthy rats produced its fibrillogenicity [35,36]. Given either it appears the activation of
measureable behavioural symptoms, likened to that of CTE in caspases is pathogenic.
humans (Table 1) [9]. It has been hypothesized that the phospho- Additionally, other studies have associated tau with increased
rylated tau found in CTE cause neuronal death in a similar mecha- cell cycle proteins, proposed to cause differentiated neurons to
nism to those found in Alzheimer’s disease (AD), as the tau re-enter the cell cycle leading to their death [37,38]. It has also
isoforms found in CTE match those of AD [10,11]. Normal tau is been suggested that tau may lead to membrane permeability
involved in stabilizing microtubule fibrils by binding to tubulin, changes, leading to mitochondrial dysfunction, raised intracellular
facilitating neurite outgrowth [12]. Ca2+ and reactive oxygen species production. This idea is based on
Immuunohistochemical studies have ascertained that phospho- the propensity for oligomers of beta amyloid and alpha-synuclin to
rylation of tau occurs following mild traumatic brain injury [13]. form pores in lipid membranes [39]. Oligomerization is a charac-
The phosphorylation of tau arises from an imbalance between teristic shared by tau. There is evidence for mitochondrial dysfunc-
kinases and phosphatase activity. The signalling pathways that tion in tauopathy mice models, indicating this theory may be
activate kinases following head trauma are yet to be ascertained, plausible, but requires more evidence for affirmation [40]. Strongly
but the levels of active extracellular signal regulated kinases 1 supported by the evidence presented is the idea that tau effects
and 2, cycline dependant kinase 5, glycogen synthase kinase 3- neurons on two levels – synaptic and axonal dysfunction and neu-
beta, protein kinase C, c-jun kinase and Akt are all increased fol- ronal loss, and this is what leads to physco-cognitive symptoms
lowing head trauma [14–19]. One if not multiple are likely are seen in CTE patients [41].
involved in the phosphorylation of tau. Hyperphosphorylation Neurons can survive for decades with NFTs [42]. By forming
causes dissociation of tau from tubulin, rendering microtubules these large fibrillary aggregates the cell is protected from the
dysfunctional. This exposes further phosphorylation sites leading immediate cytotoxic effects of oligomers, allowing to the compro-
to a state of hyperophosphorylation [20]. In this state tau is insol- mised neuron to maintain itself [43]. However, as these NFTs grow,
uble, so translocates to the neuron soma [21,22]. Accumulation of they have been reported to decrease the number of cell organelles,
insoluble tau leads to the formation of tau oligomers [21,22]. inhibit proteasome activity, and also impair anterograde axonal
Sequestration of oligomers and post translational modifications transport, all of which may negative alter cell homeostasis leading
lead to neurofibrillary tangle formation (NFT), the pathological to cell death [44–46].
hallmark of CTE [22,23]. The progressive neurodegeneration in CTE can be explained by
Whether tau oligomers or NFTs are the source of neurotoxicity the transmission of tau oligomers between neurons. In rat models
is an area of intense research. However, what is clearer is the where tau is overexpressed, it has been observed to be secreted
26 A.R. Vile, L. Atkinson / Journal of Clinical Neuroscience 41 (2017) 24–29

[47]. Likewise Kfoury et al. was able to demonstrate that both In non-CTE affected brain the neuro-destructive actions of
secretion of tau and uptake into cultured cells [48]. The notion that microglia are self-limiting, as they switch to a neuro-reparative
this applies to humans is supported by the finding of higher levels mode following activation, secreting neurotrophic factors, to repair
of tau in the cerebrospinal fluid of Alzheimer’s disease patients inflicted damage [77]. Frequent incidences of mild traumatic brain
when compared with healthy controls [49]. It is suggested that injury lead to persistent activation of microglia, not allowing them
uptake from the extracellular space is mediated by micropinocyto- to switch to neuro-reparative mode or repetition of the priming
sis. Santa-Maria et al. and Wu et al. found tau aggregates to co- effect, there is evidence for both occurring, with both leading to tis-
localise with dextran, a maker for fluid phase endocytosis, indict- sue damage [78–83]. Activation time also increases with each inci-
ing this as a possible route of uptake [50,51]. A process that is pos- dence of injury, and this leads to the chronic neuro-inflammation
sibly stimulated by the interaction between tau and heparin and tissue injury seen in CTE [78–81]. This theory has been sup-
sulfate proteoglycans, as was found by Hall et al. [52]. ported in humans by the higher incidence of translocator protein
(TSPO) levels, a marker of neuro inflammation in 9 retired NFL
players, with a history of repetitive head trauma, when compared
2.2. TAR DNA-binding protein 43 proteinopathy
with 9 age matched healthy individuals, indicating a chronic
inflammatory response [84]. Additionally, excitotoxicity is
TAR DNA-binding protein 43 (TDP-43) proteinopathy, is univer-
believed to play a role in tau hyperphosphorylation, likely through
sal by late stage CTE [2]. Overexpression of TDP-43 leads to translo-
CDK5 activation. In an in vivo mice model, kainic acid was used to
cation from the nucleus to the cytoplasm, leading to neuronal
induce excitotoxicity, and this resulted in a dramatic activation of
death [53]. Injection of TDP-43 into the cytoplasm of neurons, also
CDK5 and tau hyperphosphroylation [85]. This likely occurs
produces marked neuronal death, supporting the idea that translo-
through NMDA receptors, as NMDA receptor blockers have
cation is associated with its pathogenicity [54]. TDP-43 expression
decreased phosphorylated tau deposition [85]. The role of excitot-
is upregulated in mild traumatic brain injury, which is the under-
cicity is also supported by the correlation between areas most
lying aetiology of CTE, in addition to axotomy [55–57]. Following
effected between the significant atrophy of the hippocampus,
translocation, TDP-43 undergoes post-translational modification,
entorhinal cortex and amygdala and their higher susceptibility to
causing it to become hyperphosphorylated, ubiquinated and
excitotxicity [86]. Should the activation of kinases be proven to
cleaved, which causes aggregation [58–61].
be solely due to excitotoxins, this would represent a unifying
Numerous putative TDP-43 kinases have been identified,
mechanism in the signalling cascade, and propose a mechanism
though Casein Kinase 1d appears to be one of the more promising
by which kinases become activated following head injury. This is
of effectors, given its ability to replicate pathological phosphoryla-
yet to be determined. Additionally, there is evidence to suggest
tion of Ser 409/410 and downstream pathological alterations in cell
neuronal loss and misfolded proteins can further prime microglia
lines [62–64]. However, the relationship of these kinases to head
and this may perpetuate the disease process [87].
trauma remains to be determined.
The aforementioned is postulated to give TDP-43 a gain of func-
2.4. Diffuse axonal injury
tion toxicity, though the mechanism by which causes cell death
has not been defined [58–61]. It has also been demonstrated that
In McKee et al. systematic review of the pathological findings
loss of TDP-43 function can also lead to neuronal death, with this
CTE, axonal degeneration was found to be common and is seen
possibly being linked to its protein mis-folding and aggregation
on imaging studies of retired NFL players with CTE [2]. It is sug-
[65–67]. Cases control studies on Amyotrophic Lateral Sclerosis
gested that axonal injury suffered in mild traumatic brain injury
with cognitive impairment, have shown altered tau metabolism
is similar to that of diffuse axonal injury, just to a lesser extent
including hyperphospherylation and tau phosphatase resistance
of that is seen in severe head trauma, given there are similarities
concurrent with TDP-43 proteinopathy, suggesting TDP-43 ability
in axonal pathomorphology [88]. When the brain is subjected to
to modulate tau phosphorylation, which may be applicable to
rapid acceleration, deceleration or rotational forces, such as that
CTE [68]. However the extent to which this applies to CTE is
of mild traumatic brain injury the brain elongates and deforms,
debateable given that minimal co-localisation has been found
stretching the micro-architecture of the brain, including neurons,
[69]. Similar to tau, TDP-43 has been shown to have prion like
glial cells and blood vessels. Animal studies have demonstrated
seeding and propagation properties, with this again accounting
that areas that experience most stress are the depths of the cere-
for the spread of TDP-43 throughout the brain [70].
bral sulci and perivascularly [89,90]. The stretch forces placed on
the axolemma increases its permeability leading to calcium influx.
2.3. Microglial activation and immunoexcitoxicity Increased intracellular calcium induces calpain activation leading
to proteolysis of the axonal cytoskeleton and opening of the mito-
Mechanical trauma and disruption to the blood brain barrier, chondrial membrane permeability transition (MPT)-pore in axonal
which occurs in mild traumatic brain injury ‘prime’ microglia. To mitochondria. This leads to proapoptotic substance release, such as
date no biochemical descriptors of microglia priming have been cytochrome C, leading to further degeneration of the axon
identified, with morphological changes the only marker of micro- cytoskeletal network [91,88]. Microscopically, this is marked by
glia priming. These morphological changes have been identified mitochondrial and axonal swelling due to ATP dependant ion
in post mortem of long term survivors of traumatic brain injuries pump failure, which is seen in both mild traumatic brain injury
[71,72]. When microglia remain primed, a second incidence of mild and diffuse axonal injury [92,88]. The highest concentration of
traumatic brain injury, may fully activate the microglia, stimulat- phosphorylated tau correlates to the areas of highest areas of
ing a release of toxic levels of pro-inflammatory cytokines, stress, being the perivascular regions and depths of cerebral sulci.
chemokines, interferons, reactive oxygen species (ROS), and excito- On the basis of evidence provided in this review this may be due to
toxins; glutamate, aspartate, and QUIN [73–75]. The effects of a more pronounced activation of microglia at these sites.
cytokines involved following brain injury were reviewed by Ziebell
and Morganti-Kossmann, and were found to inherently harmful to 2.5. Amyloid beta and Lewy body pathology
the neurons [76]. Cytokines in combination with the effects of ROS
and excitotoxins lead to neurodegeneration, in what is termed A 2015 study conducted by Stein et.al. found Ab pathology in
immune excitotoxicity [74,76]. 52% of 114 neuropathologically confirmed CTE cases, with a similar
A.R. Vile, L. Atkinson / Journal of Clinical Neuroscience 41 (2017) 24–29 27

Fig. 1. Pathogenesis of CTE.

rate of 44% found by McKee et al. [2,93] When controlling for age, aggregation, with the two able to modulate each other’s aggrega-
beta amyloid pathology was of higher incidence, occurred at a tion to propagate themselves [94–97]. Such a theory also explains
younger age and at a greater rate in CTE affected patients than in higher frequency of Parkinsonism that occurs in individuals with
the general population. It is possible that the presence of Ab repre- CTE [2].
sents the development of a co-morbid Alzheimer’s disease, though
the accelerated rate that occurs in CTE would suggest that the dis- 3. Conclusion
ease process of CTE is a possible modifying factor for Ab deposition.
Additionally, there were correlates between severity of Ab pathol- The pathogenesis of the CTE is multifactorial and complex. Fig. 1
ogy and CTE stage progression. This would indicate possible inter- displays a proposed pathogenesis of CTE. Repetitive mild head
play between these factors and also a worse prognosis in those trauma has been established as a clear aetiology of the disorder.
with extensive Ab pathology. Of note, is the difference in Ab distri- This leads to microglial and kinase activation and diffuse axonal
bution in CTE compared with AD, with CTE having less neuritic pla- injury. The phosphorylation of tau, a result of kinase phosphatase
ques, but similar frequencies of diffuse plaques when compared to imbalance, induced by the head injury, leads to hyperphosphoryla-
AD. Given this CTE with Ab pathology is a likely subtype of CTE tion of tau, oligomer formation followed by sequestration into
with a faster clinical course [93]. Additionally, a-Synuclein- NFTs. The mechanism by which tau causes neuronal death is yet
positive Lewy bodies have been reported in approximately 20% of to be fully elucidated and appears to be multifactorial. Along with
CTE cases [2]. An explanation for this is provided in the evidence diffuse axonal injury and the immunoexcitotoxicity that arises
that exists for the synergism between tau and alpha-synuclein from microglial activation, phosphorylated tau leads to axonal
28 A.R. Vile, L. Atkinson / Journal of Clinical Neuroscience 41 (2017) 24–29

degeneration and neuronal death, resulting in disruption of neural [22] Cowan CM, Mudher A. Are tau aggregates toxic or protective in tauopathies?
Front Neurol 2013;4(1):114–8. http://dx.doi.org/10.3389/fneur.2013.00114.
circuitry. The disruption to neural circuitry is what underpins the
[23] Whittington RA, Bretteville A, Dickler MF, et al. Anesthesia and tau pathology.
broad psycho-cognitive abnormalities seen in CTE (Table 1). Amy- Prog Neuropsychopharmacol Biol Psychiatry 2013;4(1):147–55. http://dx.doi.
loid beta and Lewy bodies possibly modify factors to the diseases org/10.1016/j.pnpbp.2013.03.004.
course. Currently, there is no treatment for CTE. Given the inter- [24] Goedert M, Spillantini MG. A century of Alzheimer’s disease. Science 2006;314
(5800):777–81. http://dx.doi.org/10.1126/science.1132814.
relationship between phosphorylation of tau and other mecha- [25] Gendron TF, Petrucelli L. The role of tau in neurodegeneration. Mol
nisms, kinase inhibition presents itself as a possible treatment Neurodegener 2009;4:13. http://dx.doi.org/10.1186/1750-1326-4-13.
modality, and has shown success in Alzheimer’s disease, another [26] Tomlinson BE, Blessed G, Roth M. Observations on the brains of demented old
people. J Neurol Sci 1970;11(1):205–42. doi: 10.1002/(SICI)1099-1166
tauopathy [98]. Thus, this review has detailed the pathogenesis (199708)12:8<785::AID-GPS642>3.0.CO;2-F.
of CTE, as well as linking the pathophysiology to the distribution [27] Alafuzoff I, Iqbal K, Friden H, et al. Histopathological criteria for progressive
of pathological hallmarks, allowing future studies to utilise this dementia disorders: clinical-pathological correlation and classification by
multivariate data analysis. Acta Neuropathol 1987;74(1):209–25. http://dx.
knowledge for the conception of treatment and diagnostic doi.org/10.1007/BF00688184.
modalities. [28] Arriagada PV, Growdon JH, Hedley-Whyte ET, et al. senile plaques parallel
duration and severity of Alzheimer’s disease. Neurology 1992(1);42:631–639.
doi: http://dx./doi./org//10./1212/WNL.42.3.631.
References [29] Crespo-Biel N, Theunis C, Van Leuven F. Protein tau: prime cause of synaptic
and neuronal degeneration in Alzheimer’s disease. Int J Alzheimers Dis
[1] Martland HS. Punch drunk. J Am Med Assoc 1928;91(15):1103–7. http://dx. 2012;2012(3):251426. http://dx.doi.org/10.1155/2012/251426.
doi.org/10.1001/jama.1928.02700150029009. [30] Ward SM, Himmelstein DS, Lancia JK, et al. Tau oligomers and tau toxicity in
[2] McKee AC, Stern RA, Nowinski CJ, et al. The spectrum of disease in chronic neurodegenerative disease. Biochem Soc Trans 2012;40(4):667–71. http://dx.
traumatic encephalopathy. Brain 2013;136(1):43–64. http://dx.doi.org/ doi.org/10.1042/BST20120134.
10.1093/brain/aws307. [31] Su JH, Deng G, Cotman CW. Transneuronal degeneration in the spread of
[3] Ban VS, Madden CJ, Bailes JE, et al. The science and questions surrounding Alzheimer’s disease pathology: immunohistochemical evidence for the
chronic traumatic encephalopathy. Neurosurg Focus 2016;40(4):E15. http:// transmission of tau hyperphosphorylation. Neurobiology 1974;5:365–75.
dx.doi.org/10.3171/2016.2.focus15609. http://dx.doi.org/10.1006/nbdi.1997.0164.
[4] Hof PR, Bouras C, Buee L, et al. Differential distribution of neurofibrillary [32] Rohn TT et al. Caspase-9 activation and caspase cleavage of tau in the
tangles in the cerebral cort ex of dementia pugilistica and Alzheimer’s disease Alzheimer’s disease brain. Neurobiol Dis 2002;11(2):341–54. http://dx.doi.
cases. Acta Neuropathol 1992;85(1):23–30. org/10.1006/nbdi.2002.0549.
[5] Brandenburg W, Hallervorden J. Dementia pugilistica with anatomical [33] Gamblin TC et al. Caspase cleavage of tau: linking amyloid and neurofibrillary
findings. Virchows Arch 1954;325(1):680–709. tangles in Alzheimer’s disease. Proc Natl Acad Sci USA 2003;100(17):10032–7.
[6] Courville CB. Punch drunk. Its pathogenesis and pathology on the basis of a http://dx.doi.org/10.1073/pnas.1630428100.
verified case. Bull Los Angel Neuro Soc 1962;27(1):160–8. [34] Fasulo L, Ugolini G, Cattaneo A. Apoptotic effect of caspase-3 cleaved tau in
[7] Grahmann H, Ule G. Diagnosis of chronic cerebral symptoms in boxers hippocampal neurons and its potentiation by tau FTDP-mutation N279K. J
(dementia pugilistica & traumatic encephalopathy of boxers. Psychiatr Neurol Alzheimers Dis 2005;7(1):3–13. <http://www.j-alz.com/issues/7/vol7-1.html>
1956;134(1):261–83. http://dx.doi.org/10.1159/000138743. [accessed 29.03.2016].
[8] Philip H, Montenigro PH, Bernick C, et al. Clinical features of repetitive [35] Allen B, Ingram E, Takao M, et al. Abundant tau filaments and nonapoptotic
traumatic brain injury and chronic traumatic encephalopathy. Brain Pathol neurodegeneration in transgenic mice expressing human P301S Tau protein. J
2015;25(3):304–17. http://dx.doi.org/10.1111/bpa.12250. Neurosci 2002;22(21):9340–51. <http://www.jneurosci.org/content/22/21/
[9] Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, et al. Alzheimer brain- 9340.long> [accessed 01.04.2016].
derived tau oligomers propagate pathology from endogenous tau. Sci Rep [36] Spires-Jones TL, de Calignon A, Matsui T, et al. In vivo imaging reveals
2012;2(1):700. http://dx.doi.org/10.1038/srep00700. dissociation between caspase activation and acute neuronal death in tangle-
[10] McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in bearing neurons. J Neurosci 2008;28(4):862–7. http://dx.doi.org/10.1523/
athletes: progressive tautopathy after repetitive head injury. J Neuropathol JNEUROSCI.3072-08.2008.
Exp Neurol 2009;68(7):709–35. http://dx.doi.org/10.1097/ [37] Andorfer C et al. Cell-cycle reentry and cell death in transgenic mice
NEN.0b013e3181a9d503. expressing nonmutant human tau isoforms. J Neurosci 2005;25
[11] Schmidt ML, Zhukareva V, Newell KL, et al. Tau isoform profile and (22):5446–54. http://dx.doi.org/10.1523/JNEUROSCI.4637-04.2005.
phosphorylation state in dementia pugilistica recapitulate Alzheimer’s [38] Schindowski K, Belarbi K, Bretteville A, et al. Neurogenesis and cell cycle-
disease. Acta Neuropathol 2001;101(5):518–24. http://dx.doi.org/10.1007/ reactivated neuronal death during pathogenic tau aggregation. Genes Brain
s004010000330. Behav 2008;7(1):92–100. http://dx.doi.org/10.1111/j.1601-183X.2007.00377.
[12] Iqbal K, Gong CX, Liu F. Hyperphosphorylation–induced tau oligomers. Front x.
Neurol 2013;4(1):112–8. http://dx.doi.org/10.3389/fneur.2013.00112. [39] Bucciantini M, Giannoni E, Chiti F, et al. Inherent toxicity of aggregates implies
[13] Hawkins BE, Krishnamurthy S, Castillo-Carranza DL, et al. Rapid accumulation a common mechanism for protein misfolding diseases. Nature 2002;416
of endogenous tau oligomers in a rat model of traumatic brain injury possible (6880):507–11. http://dx.doi.org/10.1038/416507a.
link between traumatic brain injury and sporadic tauopathies. J Biol Chem [40] David DC, Hauptmann S, Scherping I, et al. Proteomic and functional analyses
2013;288(1):17042–50. http://dx.doi.org/10.1074/jbc.M113.472746. reveal a mitochondrial dysfunction in P301L Tau transgenic mice. J Biol Chem
[14] Kuo JR, Cheng YH, Chen YS, et al. Involvement of extracellular signal regulated 2005;280(25):23802–14. <http://www.ncbi.nlm.nih.gov/pubmed/15831501>
kinases in traumatic brain injury-induced depression in rodents. J [accessed 26.03.2016].
Neurotrauma 2013;30(14):1223–31. http://dx.doi.org/10.1089/ [41] Di J, Cohen LS, Corbo CP, et al. Abnormal tau induces cognitive impairment
neu.2012.2689. through two different mechanisms: synaptic dysfunction and neuronal loss.
[15] Li A, Zou F, Fu H, et al. Upregulation of CRM1 relates to neuronal apoptosis Sci Rep 2016;6:20833. http://dx.doi.org/10.1038/srep20833.
after traumatic brain injury in adult rats. J Mol Neurosci 2013;51(1):208–18. [42] Morsch R, Simon W, Coleman PD. Neurons may live for decades with
http://dx.doi.org/10.1007/s12031-013-9994-7. neurofibrillary tangles. J Neuropathol Exp Neurol 1999;58:188–97. <http://
[16] Shapira M, Licht A, Milman A, et al. Role of glycogen synthase kinase-3b in www.ncbi.nlm.nih.gov/pubmed/10029101. [accessed 18.03.2016]>.
early depressive behaviour induced by mild traumatic brain injury. Mol Cell [43] Congdon EE, Duff KE. Is tau aggregation toxic or protective? J. Alzheimer’s Dis.
Neurosci 2007;34(4):571–7. http://dx.doi.org/10.1016/j.mcn.2006.12.006. 2008;14(4):453–7. <https://www.researchgate.net/publication/232551940_
[17] Zohar O, Lavy R, Zi X, et al. PKC activator therapeutic for mild traumatic brain Is_tau_aggregation_toxic_or_protective> [accessed 16.03.2016].
injury in mice. Neurobiol Dis 2011;41(2):329–37. http://dx.doi.org/10.1016/j. [44] Lin WL, Lewis J, Yen SH, et al. Ultrastructural neuronal pathology in transgenic
nbd.2010.10.001. mice expressing mutant (P301L) human tau. J Neurocytol 2003;32
[18] Tran HT, Sanchez L, Brody DL. Inhibition of JNK by a peptide inhibitor reduces (9):1091–105. http://dx.doi.org/10.1023/B:NEUR.0000021904.61387.95.
traumatic brain injury-induced tauopathy in transgenic mice. J Neuropathol [45] Keck S, Nitsch R, Grune T, et al. Proteasome inhibition by paired helical
Exp Neurol 2012;71(2):116–29. http://dx.doi.org/10.1097/ filament-tau in brains of patients with Alzheimer’s disease. J Neurochem
NEN.0b013e3182456aed. 2003;85:115–22. http://dx.doi.org/10.1046/j.1471-4159.2003.01642.x.
[19] Ksiezak-Reding H, Pyo HK, Feinstein B, et al. Akt/PKB kinase phosphorylates [46] Lapointe NE, Morfini G, Pigino G, et al. The amino terminus of tau inhibits
separately Thr212 and Ser214 of tau protein in vitro. Biochim Biophys Acta kinesin-dependent axonal transport: implications for filament toxicity. J
2013;1639(3):159–68. http://dx.doi.org/10.1016/j.bbadis.2003.09.001. Neurosci Res 2009;87(2):440–51. http://dx.doi.org/10.1002/jnr.21850.
[20] Walker KR, Tesco G. Molecular mechanisms of cognitive dysfunction following [47] Barten DM, Cadelina GW, Hoque N, et al. Tau transgenic mice as models for
traumatic brain injury. Front Aging Neurosci 2013;5(1):29–43. http://dx.doi. cerebrospinal fluid tau biomarkers. J Alzheimers Dis 2011;24(2):127–41.
org/10.3389/fnagi.2013.00029. http://dx.doi.org/10.3233/JAD-2011-110161.
[21] Iqbal K, Gong CX, Liu F. Hyperphosphorylation–induced tau oligomers. Front [48] Kfoury N, Holmes BB, Jiang H, et al. Trans-cellular propagation of tau
Neurol 2013;4(1):112–8. http://dx.doi.org/10.1016/j. aggregation by fibrillar species. J Biol Chem 2012;287(23):19440–51. http://
neurobiolaging.2007.02.029. dx.doi.org/10.1074/jbc.M112.346072.
A.R. Vile, L. Atkinson / Journal of Clinical Neuroscience 41 (2017) 24–29 29

[49] Hampel H, Blennow K, Shaw LM, et al. Total and phosphorylated tau protein as [73] Perry HV, Holmes C. Microglial priming in neurodegenerative disease. Nat Rev
biological markers of Alzheimer’s disease. Exp Gerontol 2010;45(1):30–40. Neurol 2014;10(1):217–24. http://dx.doi.org/10.1038/nrneurol.2014.38.
http://dx.doi.org/10.3233/JAD-2011-110161. [74] Johnson VE, Stewart JE, Begbie FD, et al. Inflammation and white matter
[50] Santa-Maria I, Varghese M, Ksiezak-Reding H, et al. Paired helical filaments degeneration persist for years after a single traumatic brain injury. Brain
from Alzheimer disease brain induce intracellular accumulation of Tau protein 2013;136(1):28–42. http://dx.doi.org/10.1093/brain/aws322.
in aggresomes. J Biol Chem 2012;287:20522–33. http://dx.doi.org/10.1074/ [75] Combrinck MI, Perry VH, Cunningham C. Peripheral infection evokes
jbc.M111.323279. exaggerated sickness behavior in pre-clinical murine prion disease.
[51] Wu JW, Herman M, Liu L, et al. Small misfolded Tau species are internalized via Neuroscience 2002;112:7–11.
bulk endocytosis and anterogradely and retrogradely transported in neurons. J [76] Cunningham C, Wilcockson DC, Campion S, et al. Central and systemic
Biol Chem 2013;288:1856–70. http://dx.doi.org/10.1074/jbc.M112.394528. endotoxin challenges exacerbate the local inflammatory response and increase
[52] Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside neuronal death during chronic neurodegeneration. J Neurosci
to the inside of a cell. J Biol Chem 2009;284(19):12845–52. http://dx.doi.org/ 2005;25:9275–84.
10.1074/jbc.M808759200. [77] Sandhir R, Onyszchuk G, Berman NE. Exacerbated glial response in the aged
[53] Guo JL, Lee VM. Seeding of normal Tau by pathological Tau conformers drives mouse hippocampal following controlled cortical impact injury. Exp Neurol
pathogenesis of Alzheimer-like tangles. J Biol Chem 2011;286(1):15317–31. 2008;13:372–80. http://dx.doi.org/10.1016/j.expneurol.2008.06.013.
http://dx.doi.org/10.1074/jbc.M110.209296. [78] Ziebell JM, Morganti-Kossmann MC. Involvement of pro- and anti-
[54] Wu JW, Herman M, Liu L, et al. Small misfolded tau species are internalized via inflammatory cytokines and chemokines in the pathophysiology of
bulk endocytosis and anterogradely and retrogradely transported in neurons. J traumatic brain injury. Neurotherapeutics 2010;7(1):22–30. http://dx.doi.
Biol Chem 2013;288(1):1856–70. http://dx.doi.org/10.1074/jbc.M112.394528. org/10.1016/j.nurt.2009.10.016.
[55] Barmada SJ, Skibinski G, Korb E, et al. Cytoplasmic mislocalization of TDP-43 is [79] Langston JW, Fomo LS, Tetrud J, et al. Evidence of active nerve cell
toxic to neurons and enhanced by a mutation associated with familial degeneration in the substantia nigra of humans years after 1-methyl-4-
amyotrophic lateral sclerosis. J Neurosci 2010;30:639–49. http://dx.doi.org/ phenyl-1,2,3,6-tetrahydropyridine exposure. Ann Neurol 1999;46:598–605.
10.1523/JNEUROSCI.4988-09.2010. <http://www.ncbi.nlm.nih.gov/pubmed/10514096> [accessed 03.04.2016].
[56] Tatom JB, Wang D, Dayton R, et al. Mimicking aspects of frontotemporal lobar [80] Johnson VE, Stewart JE, Begbie FD, et al. Inflammation and white matter
degeneration and Lou Gehrig’s disease in rats via TDP-43 overexpression. Mol degeneration persist for years after a single traumatic brain injury. Brain
Ther 2009;17:607–13. http://dx.doi.org/10.1038/mt.2009.3. 2013;136:28–42. http://dx.doi.org/10.1093/brain/aws322.
[57] Moisse K, Mepham J, Volkening K, et al. Cytosolic TDP-43 expression following [81] Ramlackhansingh AF, Brooks DJ, Greenwood RJ, et al. Inflammation after
axotomy is associated with caspase 3 activation in NFL / mice: Support for a trauma: microglial activation and traumatic brain injury. Ann Neurol
role for TDP-43 in the physiological response to neuronal injury. Brain Res 2011;70:374–83. http://dx.doi.org/10.1002/ana.22455.
2009;1296:176–86. http://dx.doi.org/10.1016/j.brainres.2009.07.023. [82] Smith C, Gentleman SM, Leclercq PD, et al. The neuroinflammatory response in
[58] Moisse K, Mepham J, Volkening K, et al. Cytosolic TDP-43 expression following humans after traumatic brain injury. Neuropathol Appl Neurobiol
axotomy is associated with caspase 3 activation in NFL / mice: Support for a 2013;39:654–66. http://dx.doi.org/10.1111/nan.12008.
role for TDP-43 in the physiological response to neuronal injury. Brain Res [83] Gentleman SM, Leclercq PD, Moyes L, et al. Long-term intracerebral
2009;1296:176–86. http://dx.doi.org/10.1016/j.brainres.2009.07.023. inflammatory response after traumatic brain injury. Forensic Sci Int
[59] Moisse K, Volkening K, Leystra-Lantz C, et al. Divergent patterns of cytosolic 2004;146:97–104. http://dx.doi.org/10.1016/j.forsciint.2004.06.027.
TDP-43 and neuronal progranulin expression following axotomy: implications [84] Dilger RN, Johnson RW. Aging microglial cell priming, and the discordant
for TDP-43 in the physiological response to neuronal injury. Brain Res central inflammatory response to signals from the peripheral immune system.
2009;1249:202–11. http://dx.doi.org/10.1016/j.brainres.2008.10.021. J Leukoc Biol 2008;84(4):932–9. http://dx.doi.org/10.1189/jlb.0208108.
[60] Kwong LK, Neumann M, Sampathu DM, et al. TDP-43 proteinopathy: the [85] Liang Z, Liu F, Iqbal K, et al. Dysregulation of tau phosphorylation in mouse
neuropathology underlying major forms of sporadic and familial brain during excitotoxic damage. J Alzheimers Dis 2009;17(3):531–9. http://
frontotemporal lobar degeneration and motor neuron disease. Acta dx.doi.org/10.3233/JAD-2009-1069.
Neuropathol 2007;114(1):63–70. http://dx.doi.org/10.1007/s00401-007- [86] Coughlin JM, Wang Y, Munro CA. Neuroinflammation and brain atrophy in
0226-5. former NFL players: an in vivo multimodal imaging pilot study. Neurobiol Dis
[61] Buratti E, Baralle FE. Multiple roles of TDP-43 in gene expression, splicing 2015;74:58–65. http://dx.doi.org/10.1016/j.nbd.2014.10.019.
regulation, and human disease. Front. Biosci 2008;13:867–78. http://dx.doi. [87] Perry VH, Holmes C. Microglial priming in neurodegenerative disease. Nat Rev
org/10.2741/2727. Neurol 2014;10:217–24. http://dx.doi.org/10.1038/nrneurol.2014.38.
[62] Nonaka T, Suzuki G, Tanaka Y, et al. Phosphorylation of TAR DNA-binding [88] Büki A, Povlishock JT. All roads lead to disconnection? – traumatic axonal
protein of 43 kDa (TDP-43) by truncated casein kinase 1d triggers injury revisited. Acta Neurochir 2006;148(2):181–94. http://dx.doi.org/
mislocalization and accumulation of TDP-43. J Biol Chem 2016;291 10.1007/s00701-005-0674-4.
(11):5473–83. http://dx.doi.org/10.1074/jbc.M115.695379. [89] Moinuevo JL, Liado A, Rami L. Memantine: Targeting glutamate excitotoxicity
[63] Choksi DK, Roy B, Chatterjee S, et al. TDP-43 phosphorylation by casein kinase in Alzheimer’s disease and other dementias. Am J Alzheimers Dis Other Demen
Iepsilon promotes oligomerization and enhances toxicity in vivo. Hum Mol 2005;20(2):77–85. <http://aja.sagepub.com/content/20/2/77.abstract>
Gen 2014;23(4):1025–35. http://dx.doi.org/10.1093/hmg/ddt498. [accessed 27.03.2016].
[64] Nonaka T, Masai H, Hasegawa M. Phosphorylation of TDP-43 By casein kinase [90] Browne KD, Chen X, Meaney DF, et al. Mild traumatic brain injury and diffuse
1 delta facilitates mislocalization and intracellular aggregate formation of axonal injury in swine. J Neurotrauma 2011;28(9):1747–55. http://dx.doi.org/
TDP-43. Alzheimers Dement 2014;10(4):P790. http://dx.doi.org/10.1016/ 10.1089/neu.2011.1913.
j.jalz.2014.05.1533. [91] Constantinidis J, Tissot R. Generalized Alzheimer’s neurofibrillary lesions
[65] Wu LS, Cheng WC, Shen CK. Targeted depletion of TDP-43 expression in the without senile plaques. (Presentation of one anatomo-clinical case). Schweiz
spinal cord motor neurons leads to the development of amyotrophic lateral Arch Neurol Neurochir Psychiatr 1967;100(1):117–30. <https://www.
sclerosis-like phenotypes in mice. J Biol Chem 2012;287:27335–44. http://dx. researchgate.net/publication/17127397_Generalized_Alzheimer%27s_
doi.org/10.1074/jbc.M112.359000. neurofibrillary_lesions_without_senile_plaques_Presentation_of_one_
[66] Iguchi Y, Katsuno M, Niwa J, et al. Loss of TDP-43 causes age-dependent anatomo-clinical_case> [accessed 26.03.2016]..
progressive motor neuron degeneration. Brain 2013;136:1371–82. http://dx. [92] Smith DH, Chen XH, Nonaka M, et al. Accumulation of amyloid beta and tau
doi.org/10.1093/brain/awt029. and the formation of neurofilament inclusions following diffuse brain injury in
[67] Kabashi E, Lin L, Tradewell ML, et al. Gain and loss of function of ALS-related the pig. J Neuropathol Exp Neurol 1999;58(9):982–92. http://dx.doi.org/
mutations of TARDBP (TDP-43) cause motor deficits in vivo. Hum Mol Genet 10.1097/00005072-199909000-00008.
2010;19:671–83. http://dx.doi.org/10.1093/hmg/ddp534. [93] Stein TD, Montenigro PH, Alvarez VE, et al. Beta-amyloid deposition in chronic
[68] Neumann M, Tolnay M, Mackenzie IR. The molecular basis of frontotemporal traumatic encephalopathy. Acta Neuropathol 2015;130(1):21–34. http://dx.
dementia. Expert Rev Mol Med 2009;11:e23. http://dx.doi.org/10.1017/ doi.org/10.1007/s00401-015-1435-y.
S1462399409001136. [94] Geddes JW. Alpha-synuclein: a potent inducer of tau pathology. Exp Neurol
[69] McKee AC, Gavett BE, Stern RA, et al. TDP-43 proteinopathy and motor neuron 2005;192:244–50. http://dx.doi.org/10.1016/j.expneurol.2004.12.002.
disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol [95] Frasier M, Walzer M, McCarthy L, et al. Tau phosphorylation increases in
2010;69(9):918–29. http://dx.doi.org/10.1097/NEN.0b013e3181ee7d85. symptomatic mice overexpressing A30P alpha-synuclein. Exp Neurol
[70] Smethurst P, Sidle KCL, Hardy J. Review: prion-like mechanisms of transactive 2005;192:274–87. http://dx.doi.org/10.1016/j.expneurol.2004.07.016.
response DNA binding protein of 43 kDa (TDP-43) in amyotrophic lateral [96] Badiola N, de Oliveira RM, Herrera F, et al. Tau enhances a-synuclein
sclerosis (ALS). Neuropathol Appl Neurobiol 2015;41(5):578–97. http://dx.doi. aggregation and toxicity in cellular models of synucleinopathy. PLoS One
org/10.1111/nan.12206. 2011;6:e26609. http://dx.doi.org/10.1371/journal.pone.0026609.
[71] Kwong LK, Neumann M, Sampathu DM, et al. TDP-43 proteinopathy: the [97] Jellinger KA. Interaction between pathogenic proteins in neurodegenerative
neuropathology underlying major forms of sporadic and familial disorders. J Cell Mol Med 2012;16:1166–83. http://dx.doi.org/10.1111/j.1582-
frontotemporal lobar degeneration and motor neuron disease. Acta 4934.2011.01507.x.
Neuropathol 2007;114(1):63–70. http://dx.doi.org/10.1007/s00401-007- [98] Del Ser Teodoro. Phase IIa clinical trial on Alzheimer’s disease with NP12, a
0226-5. GSK3 inhibitor. Alzheimers Dement 2010;6(4):S147. http://dx.doi.org/
[72] Strong MJ, Yang W. The frontotemporal syndromes of ALS. clinicopathological 10.1016/j.jalz.2010.05.455.
correlates. J Mol Neurosci 2011;45:648–55. http://dx.doi.org/10.1007/s12031-
011-9609-0.

You might also like