Open access peer-reviewed chapter

The Neuropathology of Concussion

Written By

Ioannis Mavroudis, Ioana-Miruna Balmus, Lucian Gorgan and Alin Ciobica

Submitted: 16 May 2023 Reviewed: 05 July 2023 Published: 29 July 2023

DOI: 10.5772/intechopen.112459

From the Edited Volume

Concussion - State-of-the-Art

Edited by Ioannis Mavroudis

Chapter metrics overview

79 Chapter Downloads

View Full Metrics

Abstract

This review provides a detailed analysis of the pathophysiology involved in traumatic brain injury (TBI), with an emphasis on mild TBI and chronic traumatic encephalopathy (CTE). It explains the dynamic interaction between mechanical trauma and the neuroinflammatory response, especially the crucial role of microglia in post-TBI inflammation. Moreover, the review discusses the significance of dendritic and spinal changes as indicators of a regenerative response. The role of transactive response (TAR) DNA-binding protein 43 and tau protein in the pathogenesis of mild TBI and CTE is assessed, with tau protein changes being a potential biomarker for acute and chronic TBI-related conditions. The study also investigates syndromes commonly found in young athletes, such as second impact syndrome and juvenile head trauma syndrome. The review addresses the complex inflammatory response after mild TBI, focusing on pro-inflammatory and anti-inflammatory mediators, including IL-1, IL-6, TNF-α, and CRP as potential indicators of injury severity and outcome. The review calls for further research to elucidate the exact relationship of these factors in TBI and its long-term effects.

Keywords

  • traumatic brain injury
  • chronic traumatic encephalopathy
  • microglia
  • tau protein
  • second impact syndrome
  • inflammatory response

1. Introduction

The definition of concussion remains controversial and lacks universal agreement. The 2012 Zurich Consensus Statement on Concussion in Sport proposed that concussion and mild traumatic brain injury (TBI) should be regarded as distinct entities, and defined concussion as a “complex pathophysiological process affecting the brain” that can cause neuropathological damage. However, the statement acknowledged that concussive symptoms typically resolve spontaneously and do not produce imaging abnormalities. In contrast, recent American Academy of Neurology guidelines for sports concussion in 2013 did not differentiate between concussion and mild TBI, defining concussion as “a clinical syndrome of biomechanically induced alteration of brain function, typically affecting memory and orientation, which may involve loss of consciousness.” These guidelines noted that the terms concussion and mild TBI are often used interchangeably, highlighting a lack of consensus in their use. Therefore, concussion is currently employed in two distinct ways: (1) to describe a specific pathophysiological entity with unique diagnostic and management implications, commonly observed in the context of sports injuries; and (2) to describe a constellation of symptoms that arise after different types of TBI [1, 2].

Advertisement

2. The pathophysiology of concussion

Concussion and subconcussion injuries are caused by the acceleration and deceleration linear or rotational forces acting on the brain. This results in the elongation and deformation of the brain, causing stretching of neurons, glial cells, and blood vessels and altering membrane permeability. Although all cell compartments and blood vessels are affected by the injury, axons are especially vulnerable as they often extend long distances from the neuronal cell bodies. Axons may be injured even in the absence of the death of the neuron of origin [3, 4].

In addition to structural deformation, acceleration-deceleration forces produce a rapid release of neurotransmitters, influx of calcium, efflux of potassium, and acceleration of the cellular sodium-potassium (Na+-K+) pump to maintain membrane homeostasis, requiring large increases in glucose metabolism. These changes are referred to as the “neurometabolic cascade of concussion.”

Post-concussive hypermetabolism in the setting of decreased cerebral blood flow produces a disparity between glucose supply and demand and a cellular energy crisis [5]. Pathological studies of acute concussion and post-concussion syndrome (PCS) have shown multifocal diffuse axonal injury (DAI), microhemorrhage, astrocytosis, and perivascular clusters of activated microglia. The severity of axonal injury is generally parallel to the severity of the TBI, with mild injury producing only microscopic multifocal axonal damage and moderate and severe TBI producing more severe, widespread axonal injury. Mild TBI produces multifocal and perivascular axonal injury in the corpus callosum, fornix, subcortical white matter, and cerebellum, physical changes that may contribute to the severity of symptoms after mild TBI [3, 6]. Focal perivascular accumulations of hyperphosphorylated tau (p-tau) as neurofibrillary tangles (NFTs) and neurites and TDP-43 immunopositive neurites in the white matter have also been found after concussion, suggesting that focal axonal injury may be mechanistically associated with the development of p-tau and TDP-43 pathology. Evidence of microhemorrhage as hemosiderin and hematoidin-laden macrophages may also be present after concussion, indicating loss of microvascular integrity and breach of the blood-brain barrier following mild TBI. Structural changes in the brain after concussive injury, such as DAI and microhemorrhages, are best detected with diffusion tensor imaging (DTI) and susceptibility-weighted imaging (SWI) and are not detectable with conventional structural imaging studies, including computed tomography (CT) scan and magnetic resonance imaging (MRI) [7, 8, 9, 10].

Advertisement

3. Diffuse axonal injury

Diffuse axonal injury (DAI) is a major neuropathological consequence of TBI and is caused by the acceleration/deceleration forces that shear fragile axons during the trauma [11, 12, 13, 14]. Although DAI is more commonly seen in moderate to severe TBI, it can also occur in mild TBI, and its severity is proportional to the deceleration force [15, 16]. DAI is difficult to identify in patients with TBI using CT and conventional MRI, but novel MRI techniques, such as diffusion tensor imaging (DTI) have been found to be useful for assessing axonal integrity and identifying DAI, particularly in mild TBI patients and athletes with mild sports-related concussive or sub-concussive TBI [17, 18, 19]. Histological techniques have shown that DAI can be identified within hours after trauma while being characterized by sequential changes that begin with an acute shearing of axons, disrupted axonal transport with axonal swellings, and secondary disconnection, leading to Wallerian degeneration [11].

Diffuse axonal injury (DAI) with axolemmal disruption leads to calcium influx, neurofilament compaction, and microtubule disassembly. Calcium influx triggers microtubule disassembly, while neurofilament compaction is an early event caused by calpain-mediated proteolysis of neurofilament side arms or phosphorylation [2021]. Disruption of calcium homeostasis is the primary regulator of calpain activation, leading to increased intracellular-free calcium, and proteolytic degradation of essential cytoskeletal proteins, such as neurofilament proteins [22, 23].

Diffusion tensor imaging (DTI) is a valuable tool in diagnosing, prognosing, and managing mild TBI. DTI provides information about the microstructure and fiber tract integrity of white matter. Other techniques that may be valuable in evaluating mild TBI include alterations in brain activation through BOLD signals, resting state functional connectivity, magnetic resonance spectroscopy, and SPECT imaging.

Blast injury is becoming an increasingly important form of TBI in civilian and military populations, with the majority of injuries associated with blast exposure. Individuals exposed to blast injury are susceptible to acute and long-term neuropsychiatric and cognitive consequences. Some military veterans with a history of blast exposure show neuropathological changes of chronic traumatic encephalopathy (CTE) during autopsy, while single-blast exposure in wild-type laboratory mice produces neuropathological changes of axonal injury, neuroinflammation, microvascular injury, and abnormal tau pathology, as well as neurobehavioral abnormalities. A post-mortem series of military veterans with documented histories of blast exposure showed focal neuropathological changes of CTE, including cortical foci of perivascular tau pathology, disseminated microgliosis and astrocytosis, myelinated axonopathy, and focal neurodegeneration, very similar to mild CTE pathology found in the brains of athletes with a history of repetitive concussive injury [4]. Also, the clinical symptoms experienced by veterans with blast injury include progressive affective lability, irritability, distractibility, executive dysfunction, memory disturbances, and cognitive deficits [4].

Advertisement

4. Microtubule disorganization

Microtubule disorganization may be a direct effect of dynamic axon stretching, leading to immediate breakage and buckling of microtubules post-injury, which triggers progressive microtubule disassembly [24]. This results in the accumulation of organelles that are transported in the axon, and axonal swelling known as axonal retraction balls, leading to eventual disconnection and axotomy [20, 21]. Neuronal damage with axonal bulbs and swellings is most commonly found in the cortical sulci at the interface between gray and white matter [25]. DTI studies have shown that the extent of DAI after mild TBI is related to post-concussion cognitive problems [26].

Advertisement

5. The role of microglia

Microglia plays an essential role in the immune system in the brain and mediates the inflammatory response after TBI. Studies in animal models of TBI have shown that activated microglia migrate rapidly toward damaged tissue, forming extended cytoplasmic processes that create a potential barrier between healthy and injured tissues, indicating that microglial activation is a response to axonal damage [27, 28]. This microglial response is associated with the upregulation of both pro- and anti-inflammatory genes, chemokines, and other inflammatory mediators [29]. However, it is still not clear whether modulation of this inflammatory response to brain trauma may have any therapeutic effects. While pharmacological reduction of microglial activation might reduce inflammation and improve neuronal survival, microglial activation might stimulate axonal regeneration after injury [30].

Advertisement

6. Dendritic and spinal changes

After TBI, dendritic and synaptic sprouting occurs, leading to increased dendritic arborization and synaptogenesis as part of a regenerative response [31]. Transcription factors c-Jun and ATF-3 have been implicated in axonal regeneration after DAI [32]. Structural proteins, including growth-associated protein GAP-43, have also been associated with neurite sprouting of disconnected damaged axons after the acute phase of TBI [33].

Advertisement

7. TDP-43 deposition

TAR DNA-binding protein 43 (TDP-43) may also play a role in mild TBI and CTE pathogenesis. TDP-43 accumulation is a feature of several neurodegenerative diseases, including CTE, AD, and dementia with Lewy bodies. Recent studies have shown that TDP-43 accumulations occur in boxers and American football players with CTE after repeated brain trauma in several gray matter structures, including the brainstem, basal ganglia cortical areas, and subcortical white matter. TDP-43 accumulation after TBI may be part of a physiological injury response, and animal experiments suggest that axonal damage results in an upregulation of TDP-43 expression [34, 35, 36].

Advertisement

8. The role of tau protein

Tau protein, characterized by a molecular weight ranging between 48 and 67 kDa, serves as a crucial structural component within the axonal cytoskeleton of both the central nervous system (CNS) and peripheral nervous system (PNS) [37, 38]. This microtubule-associated protein is predominantly found in unmyelinated cortical axons, contributing significantly to their structural integrity [39, 40]. Although the expression of tau is mainly observed in the brain, it is also present in extracranial tissues, such as the liver, kidneys, and testis [41]. Historically, research investigating TBI biomarkers has primarily concentrated on total tau (T-tau). However, recent studies have expanded their focus to include phosphorylated tau (P-tau) and cleaved tau (C-tau) as well. Following a TBI, there is a noticeable increase in tau levels within cerebrospinal fluid (CSF) and plasma [42]. Elevated concentrations of tau protein in CSF have been identified in patients who have experienced TBI, with admission CSF tau levels displaying a correlation with the patients’ long-term outcomes [43, 44]. A study analyzing CSF samples from Olympic boxers within one to six days following their bouts revealed a significant increase in tau levels in the boxers’ CSF, as compared to healthy control samples [45]. Moreover, higher concentrations of CSF tau were observed in Olympic boxers post-bout, though this was not necessarily linked to the number of head impacts received [46]. Similar findings were reported in a study involving college football players where no correlation was observed between the number of mild TBIs or concussions and tau concentrations. Increased plasma tau levels were also noted following training sessions [47].

In chronic neurodegenerative disorders, such as Alzheimer’s disease (AD), CSF T-tau concentrations exhibit a weak correlation with plasma T-tau concentrations [48]. In contrast, a stronger correlation more likely exists in cases of acute TBI, contingent upon injury severity and the subsequent release of T-tau from neurons into both CSF and plasma. Notably, the elevation of tau levels post-TBI tends to persist longer in CSF (weeks) compared to blood (days) [49]. Given the typically low concentrations of tau protein in peripheral blood during both healthy and diseased states, accurate measurement through conventional immunoassays has proven challenging. The advent of ultrasensitive Single molecule array (Simoa) technology has facilitated the precise quantification of T-tau in both plasma and serum [50]. Multiple studies have documented elevated plasma T-tau concentrations in relation to TBI [51], with tau levels generally peaking between 12 and 24 hours after the injury and occasionally persisting at high levels.

The previous studies suggested that tau levels increases following TBI may exhibit both acute and chronic trajectories. While the initial increase in tau levels is indicative of acute neuronal damage, a secondary increase may be associated with chronic neurodegenerative processes and secondary pathologies [52]. Notably, blood tau concentrations have been observed to rise with age [53], and recent findings have reported distinct temporal profiles and substantially higher T-tau levels in female athletes with concussions, as compared to their male counterparts [54]. Furthermore, significant correlations have been found between serum tau concentrations and neurological outcomes in patients who have experienced resuscitated cardiac arrest [55].

In studies involving professional ice hockey players with concussions, plasma T-tau concentrations were elevated one hour post-injury compared to pre-season levels, and accurately predicted return-to-play (RTP) time [56]. Additionally, a study focusing on concussed athletes found that plasma T-tau concentrations six hours after injury correlated significantly with RTP time [57]. Elevated plasma tau levels have also been reported in military personnel exposed to blast injuries within the prior 18 months [50], with higher exosomal tau concentrations being associated with chronic symptoms in military personnel after mild TBI [58]. In a study encompassing TBIs of varying severity, plasma T-tau concentrations successfully differentiated mild TBI cases from controls when samples were collected within 24 hours of injury [59]. Serum tau levels have similarly been identified as significant outcome predictors following TBI [60]. Recent research has demonstrated that acute plasma P-tau concentrations and the P-tau/T-tau ratio outperform T-tau concentrations in predicting TBI outcomes [61]. However, plasma T-tau concentrations upon admission were unable to distinguish between incomplete and complete recovery in cases of single and uncomplicated mild TBIs [62]. Conflicting results have been reported concerning the utilization of C-tau as a fluid biomarker for the acute biochemical diagnosis of mild TBI [62, 63].

Advertisement

9. Second impact syndrome and juvenile head trauma syndrome

The second impact syndrome (SIS) and juvenile head trauma syndrome are conditions that affect children and young adults who have suffered minor brain trauma. Juvenile head trauma syndrome refers to the catastrophic or fatal cerebral edema and coma that can result from a single injury in this population. SIS occurs when an athlete experiences a mild head injury or concussion, then suffers a second head injury before the symptoms associated with the first injury have resolved, producing rapid cerebral swelling. SIS typically affects young athletes, particularly males, ranging in age from 10 to 24 years, with a mean age of 17.9 years. Most athletes reported to have SIS were American football players, usually at the high school level, but it has also been reported in association with boxing, karate, skiing, and ice hockey. SIS is thought to result from an abrupt post-traumatic loss of cerebral blood flow auto-regulation and catecholamine release that create a rapid increase in intracranial blood volume and catastrophic cerebral edema. In two-thirds of cases, a thin, acute subdural hematoma has been found on neuroimaging or at autopsy, which may reflect the hyperemic state, in the absence of other major hematomas or space-occupying lesions. The relationship of SIS with juvenile head trauma syndrome or with malignant cerebral edema after mild TBI is uncertain, and both may be manifestations of the same underlying pathophysiology [64].

Advertisement

10. The inflammatory response in mild TBI

The inflammatory response is a multifaceted process that involves the activation of various cell types, including microglia and astrocytes, and the release of a multitude of pro-inflammatory and anti-inflammatory mediators [65]. Numerous studies have demonstrated that the inflammatory response is activated early after mild TBI and can persist for several weeks or even months. Inflammatory biomarkers, such as interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and C-reactive protein (CRP), have been proposed as potential indicators of mild TBI [20, 66, 67]. These biomarkers are elevated in individuals with mild TBI, and their levels have been linked to the severity and outcome of the injury [68]. Inflammation is characterized by the activation of immune cells and the release of inflammatory mediators, such as cytokines and chemokines [69]. In the context of concussion, inflammation has been proposed as a potential contributor to the pathophysiology of the injury and the persistence of symptoms in some individuals [70]. Several studies have investigated the levels of inflammatory biomarkers in individuals with concussions. Some studies have found elevated levels of cytokines, such as IL-6 and TNF-α, in the serum or CSF of individuals affected by concussion events, while others have not observed significant differences in inflammatory biomarker levels between individuals with trauma and controls [71, 72, 73].

References

  1. 1. McCrory P, Meeuwisse WH, Aubry M, et al. Consensus statement on concussion in sport: The 4th international conference on concussion in sport held in Zurich, November 2012. British Journal of Sports Medicine. 2013;47:250-258
  2. 2. Giza CC, Kutcher JS, Ashwal S, et al. Summary of evidence-based guideline update: Evaluation and management of concussion in sports: Report of the guideline development subcommittee of the American Academy of Neurology. Neurology. 2013;80:2250-2257
  3. 3. Blennow K, Hardy J, Zetterberg H. The neuropathology and neurobiology of traumatic brain injury. Neuron. 2012;76(5):886-899. DOI: 10.1016/j.neuron.2012.11.021
  4. 4. Mavroudis I, Kazis D, Chowdhury R, Petridis F, Costa V, Balmus IM, et al. Post-concussion syndrome and chronic traumatic encephalopathy: Narrative review on the neuropathology, neuroimaging and fluid biomarkers. Diagnostics (Basel). 2022;12(3):740. DOI: 10.3390/diagnostics12030740
  5. 5. Graham DI, Gennarelli TA, McIntosh TK. Trauma. In: Graham DI, Lantos PL, editors. Greenfield's Neuropathology. London: Arnold; 2002. pp. 823-898
  6. 6. Zidan M, Jesser J, Herweh C, Jost J, Heiland S, Meyding-Lamadé U, et al. Deep grey matter volume is reduced in amateur boxers as compared to healthy age-matched controls. Clinical Neuroradiology. Jun 2023;33(2):475-482. doi: 10.1007/s00062-022-01233-3. Epub 2022 Dec 16. PMID: 36525030; PMCID: PMC10220131
  7. 7. McKee AC, Robinson ME. Military-related traumatic brain injury and neurodegeneration. Alzheimer's & Dementia. 2014;10(3 Suppl):S242-S253
  8. 8. McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in athletes: Progressive tauopathy after repetitive head injury. Journal of Neuropathology and Experimental Neurology. 2009;68:709-735
  9. 9. Huang MX, Theilmann RJ, Robb A, et al. Integrated imaging approach with MEG and DTI to detect mild traumatic brain injury in military and civilian patients. Journal of Neurotrauma. 2009;26:1213-1226
  10. 10. Wilde EA, McCauley SR, Hunter JV, et al. Diffusion tensor imaging of acute mild traumatic brain injury in adolescents. Neurology. 2008;70:948-955
  11. 11. Johnson VE, Stewart W, Smith DH. Axonal pathology in traumatic brain injury. Experimental Neurology. 2013;246:35-43
  12. 12. Meaney DF, Smith DH, Shreiber DI, et al. Biomechanical analysis of experimental diffuse axonal injury. Journal of Neurotrauma. 1995;12:689-694
  13. 13. Adams JH, Doyle D, Ford I, Gennarelli TA, Graham DI, McLellan DR. Diffuse axonal injury in head injury: Definition, diagnosis and grading. Histopathology. 1989 Jul;15(1):49-59. DOI: 10.1111/j.1365-2559.1989.tb03040.x
  14. 14. Alexander MP. Mild traumatic brain injury: Pathophysiology, natural history, and clinical management. Neurology. 1995;45(7):1253-1260. DOI: 10.1212/wnl.45.7.1253
  15. 15. Oppenheimer DR. Microscopic lesions in the brain following head injury. Journal of Neurology, Neurosurgery, and Psychiatry. 1968;31(4):299-306. DOI: 10.1136/jnnp.31.4.299
  16. 16. Elson LM, Ward CC. Mechanisms and pathophysiology of mild head injury. Seminars in Neurology. 1994;14(1):8-18. DOI: 10.1055/s-2008-1041053
  17. 17. Bazarian JJ, Zhong J, Blyth B, Zhu T, Kavcic V, Peterson D. Diffusion tensor imaging detects clinically important axonal damage after mild traumatic brain injury: A pilot study. Journal of Neurotrauma. 2007;24(9):1447-1459. DOI: 10.1089/neu.2007.0241
  18. 18. Mayer AR, Ling J, Mannell MV, Gasparovic C, Phillips JP, Doezema D, et al. A prospective diffusion tensor imaging study in mild traumatic brain injury. Neurology. 2010;74(8):643-650. DOI: 10.1212/WNL.0b013e3181d0ccdd
  19. 19. Bazarian JJ, Zhu T, Blyth B, Borrino A, Zhong J. Subject-specific changes in brain white matter on diffusion tensor imaging after sports-related concussion. Magnetic Resonance Imaging. 2012;30(2):171-180. DOI: 10.1016/j.mri.2011.10.001
  20. 20. Giza CC, Hovda DA. The neurometabolic cascade of concussion. Journal of Athletic Training. 2001;36(3):228-235
  21. 21. Barkhoudarian G, Hovda DA, Giza CC. The molecular pathophysiology of concussive brain injury. Clinics in Sports Medicine. 2011;30(1):33-48, vii–iii. DOI: 10.1016/j.csm.2010.09.001
  22. 22. McCracken E, Hunter AJ, Patel S, Graham DI, Dewar D. Calpain activation and cytoskeletal protein breakdown in the corpus callosum of head-injured patients. Journal of Neurotrauma. 1999;16(9):749-761. DOI: 10.1089/neu.1999.16.749
  23. 23. Saatman KE, Creed J, Raghupathi R. Calpain as a therapeutic target in traumatic brain injury. Neurotherapeutics. 2010;7(1):31-42. DOI: 10.1016/j.nurt.2009.11.002
  24. 24. Tang-Schomer MD, Patel AR, Baas PW, Smith DH. Mechanical breaking of microtubules in axons during dynamic stretch injury underlies delayed elasticity, microtubule disassembly, and axon degeneration. The FASEB Journal. 2010;24(5):1401-1410. DOI: 10.1096/fj.09-142844
  25. 25. Chen XH, Siman R, Iwata A, Meaney DF, Trojanowski JQ , Smith DH. Long-term accumulation of amyloid-beta, beta-secretase, presenilin-1, and caspase-3 in damaged axons following brain trauma. The American Journal of Pathology. 2004;165(2):357-371. DOI: 10.1016/s0002-9440(10)63303-2
  26. 26. Lipton ML, Gellella E, Lo C, Gold T, Ardekani BA, Shifteh K, et al. Multifocal white matter ultrastructural abnormalities in mild traumatic brain injury with cognitive disability: A voxel-wise analysis of diffusion tensor imaging. Journal of Neurotrauma. 2008;25(11):1335-1342. DOI: 10.1089/neu.2008.0547
  27. 27. Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nature Neuroscience. 2005;8(6):752-758. DOI: 10.1038/nn1472
  28. 28. Shitaka Y, Tran HT, Bennett RE, Sanchez L, Levy MA, Dikranian K, et al. Repetitive closed-skull traumatic brain injury in mice causes persistent multifocal axonal injury and microglial reactivity. Journal of Neuropathology and Experimental Neurology. 2011;70(7):551-567. DOI: 10.1097/NEN.0b013e31821f891f
  29. 29. 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. DOI: 10.1016/j.nurt.2009.10.016
  30. 30. Loane DJ, Byrnes KR. Role of microglia in neurotrauma. Neurotherapeutics. 2010;7(4):366-377. DOI: 10.1016/j.nurt.2010.07.002
  31. 31. Keyvani K, Schallert T. Plasticity-associated molecular and structural events in the injured brain. Journal of Neuropathology and Experimental Neurology. 2002;61(10):831-840. DOI: 10.1093/jnen/61.10.831
  32. 32. Greer JE, McGinn MJ, Povlishock JT. Diffuse traumatic axonal injury in the mouse induces atrophy, c-Jun activation, and axonal outgrowth in the axotomized neuronal population. The Journal of Neuroscience. 2011;31(13):5089-5105. DOI: 10.1523/JNEUROSCI.5103-10.2011
  33. 33. Christman CW, Salvant JB Jr, Walker SA, Povlishock JT. Characterization of a prolonged regenerative attempt by diffusely injured axons following traumatic brain injury in adult cat: A light and electron microscopic immunocytochemical study. Acta Neuropathologica. 1997;94(4):329-337. DOI: 10.1007/s004010050715
  34. 34. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314(5796):130-133. DOI: 10.1126/science.1134108
  35. 35. Kadokura A, Yamazaki T, Lemere CA, Takatama M, Okamoto K. Regional distribution of TDP-43 inclusions in Alzheimer disease (AD) brains: Their relation to AD common pathology. Neuropathology. 2009;29(5):566-573. DOI: 10.1111/j.1440-1789.2009.01017.x
  36. 36. King A, Sweeney F, Bodi I, Troakes C, Maekawa S, Al-Sarraj S. Abnormal TDP-43 expression is identified in the neocortex in cases of dementia pugilistica, but is mainly confined to the limbic system when identified in high and moderate stages of Alzheimer's disease. Neuropathology. 2010;30(4):408-419. DOI: 10.1111/j.1440-1789.2009.01085.x
  37. 37. Olivera A, Lejbman N, Jeromin A, et al. Peripheral Total tau in military personnel who sustain traumatic brain injuries during deployment. JAMA Neurology. 2015;72:1109
  38. 38. Rubenstein R, Chang B, Davies P, Wagner AK, Robertson CS, Wang KKW. A novel, ultrasensitive assay for tau: Potential for assessing traumatic brain injury in tissues and biofluids. Journal of Neurotrauma. 2015;32:342-352
  39. 39. Hanes J, Zilka N, Bartkova M, Caletkova M, Dobrota D, Novak M. Rat tau proteome consists of six tau isoforms: Implication for animal models of human tauopathies. Journal of Neurochemistry. 2009;108:1167-1176
  40. 40. Zetterberg H, Smith DH, Blennow K. Biomarkers of mild traumatic brain injury in cerebrospinal fluid and blood. Nature Reviews. Neurology. 2013;9:201-210
  41. 41. Morris M, Maeda S, Vossel K, Mucke L. The many faces of tau. Neuron. 2011;70:410-426
  42. 42. Shahim P, Gill JM, Blennow K, Zetterberg H. Fluid biomarkers for chronic traumatic encephalopathy. Seminars in Neurology. 2020;40:411-419
  43. 43. Ost M, Nylén K, Csajbok L, et al. Initial CSF total tau correlates with 1-year outcome in patients with traumatic brain injury. Neurology. 2006;67:1600-1604
  44. 44. Franz G, Beer R, Kampfl A, et al. Amyloid beta 1-42 and tau in cerebrospinal fluid after severe traumatic brain injury. Neurology. 2003;60:1457-1461
  45. 45. Neselius S, Brisby H, Theodorsson A, Blennow K, Zetterberg H, Marcusson J. CSF-biomarkers in Olympic boxing: Diagnosis and effects of repetitive head trauma. PLoS One. 2012;7:e33606
  46. 46. Zetterberg H, Hietala MA, Jonsson M, et al. Neurochemical aftermath of amateur boxing. Archives of Neurology. 2006;63:1277-1280
  47. 47. Kawata K, Liu CY, Merkel SF, Ramirez SH, Tierney RT, Langford D. Blood biomarkers for brain injury: What are we measuring? Neuroscience and Biobehavioral Reviews. 2016;68:460-473
  48. 48. Mattsson N, Zetterberg H, Janelidze S, et al. Plasma tau in Alzheimer disease. Neurology. 2016;87:1827-1835
  49. 49. Ledreux A, Pryhoda MK, Gorgens K, et al. Assessment of long-term effects of sports-related concussions: Biological mechanisms and exosomal biomarkers. Frontiers in Neuroscience. 2020;14:761
  50. 50. Hossain I, Mohammadian M, Takala RSK, et al. Admission levels of total tau and β-amyloid isoforms 1-40 and 1-42 in predicting the outcome of mild traumatic brain injury. Frontiers in Neurology. 2020;11:325
  51. 51. Rubenstein R, Chang B, Yue JK, et al. Comparing plasma phospho tau, total tau, and phospho tau–total tau ratio as acute and chronic traumatic brain injury biomarkers. JAMA Neurology. 2017;74:1063
  52. 52. Walker A, Chapin B, Abisambra J, DeKosky ST. Association between single moderate to severe traumatic brain injury and long-term tauopathy in humans and preclinical animal models: A systematic narrative review of the literature. Acta Neuropathologica Communications. 2022;10:1-20
  53. 53. Blomberg M, Jensen M, Basun H, Lannfelt L, Wahlund LO. Cerebrospinal fluid tau levels increase with age in healthy individuals. Dementia and Geriatric Cognitive Disorders. 2001;12:127-132
  54. 54. Mondello S, Guedes VA, Lai C, Jeromin A, Bazarian JJ, Gill JM. Sex differences in circulating T-tau trajectories after sports-concussion and correlation with outcome. Frontiers in Neurology. 2020;11:651
  55. 55. Randall J, Mörtberg E, Provuncher GK, et al. Tau proteins in serum predict neurological outcome after hypoxic brain injury from cardiac arrest: Results of a pilot study. Resuscitation. 2013;84:351-356
  56. 56. Shahim P, Tegner Y, Wilson DH, et al. Blood biomarkers for brain injury in concussed professional ice hockey players. JAMA Neurology. 2014;71:684
  57. 57. Shahim P, Tegner Y, Marklund N, Blennow K, Zetterberg H. Neurofilament light and tau as blood biomarkers for sports-related concussion. Neurology. 2018;90:E1780-E1788
  58. 58. Gill J, Mustapic M, Diaz-Arrastia R, et al. Higher exosomal tau, amyloid-beta 42 and IL-10 are associated with mild TBIs and chronic symptoms in military personnel. Brain Injury. 2018;32:1277-1284
  59. 59. Bogoslovsky T, Wilson D, Chen Y, et al. Increases of plasma levels of glial fibrillary acidic protein, tau, and amyloid β up to 90 days after traumatic brain injury. Journal of Neurotrauma. 2017;34:66-73
  60. 60. Liliang PC, Liang CL, Weng HC, et al. τ proteins in serum predict outcome after severe traumatic brain injury. The Journal of Surgical Research. 2010;160:302-307
  61. 61. Bazarian JJ, Zemlan FP, Mookerjee S, Stigbrand T. Serum S-100B and cleaved-tau are poor predictors of long-term outcome after mild traumatic brain injury. Brain Injury. 2006;20:759-765
  62. 62. Forouzan A, Motamed H, Delirrooyfard A, Zallaghi S. Serum cleaved tau protein and clinical outcome in patients with minor head trauma. Open Access Emergency Medicine. 2020;12:7
  63. 63. Mondello S, Sorinola A, Czeiter E, et al. Blood-based protein biomarkers for the management of traumatic brain injuries in adults presenting to emergency departments with mild brain injury: A living systematic review and meta-analysis. Journal of Neurotrauma. 2021;38:1086-1106
  64. 64. Mckee AC, Daneshvar DH. The neuropathology of traumatic brain injury. Handbook of Clinical Neurology. 2015;127:45-66. DOI: 10.1016/B978-0-444-52892-6.00004-0
  65. 65. Chiu MJ, Fan LY, Chen TF, Chen YF, Chieh JJ, Horng HE. Plasma tau levels in cognitively normal middle-aged and older adults. Frontiers in Aging Neuroscience. 2017;9:51
  66. 66. Smith GS, Schneiderman AI, Bailes JE. The epidemiology of sports-related traumatic brain injury. Current Sports Medicine Reports. 2009;8:202-207
  67. 67. Guskiewicz KM, Marshall SW, Bailes J, et al. Epidemiology of concussion in collegiate and high school football players. The American Journal of Sports Medicine. 2000;28:643-650
  68. 68. Hoge CW, McGurk D, Thomas JL, et al. Mild traumatic brain injury in U.S. soldiers returning from Iraq. The New England Journal of Medicine. 2008;358:453-463
  69. 69. Broglio SP, MacKenzie EJ, Sills AK, et al. A systematic review of high school and collegiate American football games: Player injury rate and injury risk per 1,000 athlete-exposures. The American Journal of Sports Medicine. 2009;37:519-528
  70. 70. Yang H, Chen Y, Wei L, et al. Increased serum levels of inflammatory biomarkers in patients with mild traumatic brain injury: A systematic review and meta-analysis. Journal of Neurotrauma. 2017;34:623-632
  71. 71. Kolls JK, McCray PB Jr. The immune response to respiratory viruses. Nature Reviews. Immunology. 2004;4:667-676
  72. 72. Iwamoto T, Iwamoto Y, Masumoto K, et al. Elevated serum levels of cytokines in patients with mild head injury. Brain Injury. 2004;18:421-426
  73. 73. Bazarian JJ, Biberthaler P, Welch RD, et al. Serum S100B and neuron-specific enolase and their role as markers of brain injury. Journal of Neurotrauma. 2005;22:1457-1465

Written By

Ioannis Mavroudis, Ioana-Miruna Balmus, Lucian Gorgan and Alin Ciobica

Submitted: 16 May 2023 Reviewed: 05 July 2023 Published: 29 July 2023