Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2456 2023-08-29 09:59:38 |
2 Reference format revised. Meta information modification 2456 2023-08-29 10:05:48 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Halicki, M.J.; Hind, K.; Chazot, P.L. Blood-Based Biomarkers in Chronic Traumatic Encephalopathy Diagnosis. Encyclopedia. Available online: https://encyclopedia.pub/entry/48576 (accessed on 03 May 2024).
Halicki MJ, Hind K, Chazot PL. Blood-Based Biomarkers in Chronic Traumatic Encephalopathy Diagnosis. Encyclopedia. Available at: https://encyclopedia.pub/entry/48576. Accessed May 03, 2024.
Halicki, Michal J., Karen Hind, Paul L. Chazot. "Blood-Based Biomarkers in Chronic Traumatic Encephalopathy Diagnosis" Encyclopedia, https://encyclopedia.pub/entry/48576 (accessed May 03, 2024).
Halicki, M.J., Hind, K., & Chazot, P.L. (2023, August 29). Blood-Based Biomarkers in Chronic Traumatic Encephalopathy Diagnosis. In Encyclopedia. https://encyclopedia.pub/entry/48576
Halicki, Michal J., et al. "Blood-Based Biomarkers in Chronic Traumatic Encephalopathy Diagnosis." Encyclopedia. Web. 29 August, 2023.
Blood-Based Biomarkers in Chronic Traumatic Encephalopathy Diagnosis
Edit

Chronic Traumatic Encephalopathy (CTE) is a neurodegenerative disease consistently associated with repetitive traumatic brain injuries (TBIs), which makes multiple professions, such as contact sports athletes and the military, especially susceptible to its onset. There are currently no approved biomarkers to diagnose CTE, thus it can only be confirmed through a post-mortem brain autopsy. Several imaging and cerebrospinal fluid biomarkers have shown promise in the diagnosis. However, blood-based biomarkers can be more easily obtained and quantified, increasing their clinical feasibility and potential for prophylactic use. 

biomarkers CTE neurodegeneration TBI miRNA exosomes

1. Introduction

Neurodegenerative diseases are debilitating and are increasingly more common conditions, with dementia, their major consequence, projected to affect 139 million people worldwide in 2050 [1]. However, there is currently no cure or disease-modifying treatment for dementia. Chronic Traumatic Encephalopathy (CTE), first described in former boxers as ‘punch drunk syndrome’ [2], is an example of a condition described to cause deficits such as cognitive impairment, mental disturbance, and motor symptoms [3], which are all associated with dementia. As neurodegenerative condition management focuses on decreasing the patients’ suffering and decelerating the development of the condition [4], an early medical intervention in the case of suspected CTE could increase healthy life years by delaying the onset and worsening of these debilitating symptoms [5]. Unfortunately, the lack of clinically approved biomarkers for many neurodegenerative conditions, including CTE, translates into the inability to diagnose and intervene in the diseases’ prodromal, i.e., before the onset of symptoms, or the early stage.
CTE has now been confirmed by post-mortem brain autopsies in many former contact sports players, such as hockey [6] and rugby [3][6][7][8][9][10], as well as deployed military personnel [3][9][11], with the history, frequency, and intensity of traumatic brain injury (TBI) being the only risk factors consistently linked with the occurrence of CTE [12]. Notably, McKee et al. [13] analyzed over 600 published cases of neuropathologically-confirmed CTE, concluding that 97% of those were associated with repetitive head impacts (RHI). However, the inability to diagnose CTE ante-mortem impedes prophylaxis, early diagnosis, and potential symptomatic treatment in the groups with a high TBI risk. This is further aggravated by non-specific initial symptoms of the disease that can be behavioral, cognitive, mood and motor-related [14][15]. Several studies reported increased incidence of poor mental health, sleep problems, cognitive impairment, and dementia in professions with high exposure to repetitive TBIs (rTBIs) [16][17][18][19], potentially indicating different stages of CTE, which emphasizes the need for biomarkers of the disease. While imaging [20] and cerebrospinal fluid (CSF) [21][22] biomarkers of CTE have been investigated with many yielding promising results, they are expensive and invasive techniques, respectively, thus limiting their use in prophylaxis and diagnosis. On the contrary, blood-based biomarkers can be easily and non-invasively collected and have already been investigated in conditions such as Alzheimer’s disease (AD) [23].

2. Pathology of CTE and Rationale for Blood-Based Biomarkers

CTE is a progressive tauopathy characterized by the deposition of neurofibrillary tangles (NFTs) consisting mainly of hyperphosphorylated microtubule-associated protein tau (p-tau) in the perivascular depths of cortical sulci, which increases with the progression of a disease, and is linked to trauma incurred during a TBI [24][25][26]. The aggregation of tau in NFTs induces several neurotoxic mechanisms, including microtubule destabilization, synapse loss, and potential aberrations of intracellular signaling, causing neuronal death [27]. This leads to macro-scale changes, such as brain atrophy and a consequent decrease in brain volume, in the advanced stages of the disease [28][29]. Substantial force impact on the head in a TBI causes a diffuse axonal injury (DAI), which can result in the breakage of axons and a subsequent release of axonal proteins, such as tau, into the interstitial fluid and the CSF [30][31][32][33]. On the other hand, TBI also increases the permeability of the blood–brain barrier (BBB) [34] leading to a possible efflux of axonal proteins into the systemic circulation (Figure 1). This two-way pathological mechanism allows for the detection and quantification of biomarkers from blood samples [35]. An ideal blood-based biomarker should be diagnostically accurate, i.e., be able to correctly discern between patients suffering from CTE and patients who are not; selective towards CTE, i.e., be able to discern CTE from other tauopathies, for example, AD; and feasible, i.e., be easily detectable and quantifiable from blood [36][37].
Figure 1. Impairment of the blood–brain barrier (BBB) following a traumatic brain injury (TBI) and a subsequent outflow of biomarkers into the systemic circulation. TBI damages axons and instigates an inflammatory response leading to efflux of neurodegenerative biomarkers, such as phosphorylated tau (p-tau) from neurons, as well as inflammation biomarkers, such as glial fibrillary acidic protein (GFAP) from astrocyte endfeet, into interstitial fluid (ISF). TBI can also damage cells maintaining the BBB (damaged cells in dark red), leading to increased permeability of the BBB and, thus, the influx of biomarkers into blood capillaries. Created with biorender.com (accessed on 20 June 2023).
Based on the molecular pathology of CTE, two main groups of potential biomarkers can be distinguished. Potential biomarkers of neurodegeneration indicate a long-term neuronal injury and include, for instance, total (t-tau) or phosphorylated tau (p-tau). Potential biomarkers of neuroinflammation, in turn, can include glial fibrillary acidic protein (GFAP) and inflammatory cytokines, which indicate the immune activation in the central nervous system (CNS) through processes such as microgliosis or astrogliosis [38][39][40][41][42].

3. Biomarkers of Neurodegeneration in CTE

3.1. Total Tau and Phosphorylated Tau

Tau plays the most prominent role in the pathology of CTE, through the formation of NFTs and consequent neuronal dysfunction and death [27]. Numerous studies have shown a significant elevation in the extracellular p-tau deposition in the brains of people with autopsy-confirmed CTE, compared to healthy controls (e.g., Johnson et al. [43]). Most studies on the role of tau in the diagnosis of the long-term effects of TBI have focused on t-tau detectable from blood, which corresponds to compromised BBB, and p-tau, in line with signaling compromised BBB, which indicates the presence of DAI and neurotoxic mechanisms [24][25][26]. Thus far, tau has yielded promising results as an imaging biomarker [44][45], but mixed results as a biomarker from CSF [21][22][46][47]. However, both forms were shown to be significantly elevated in the brain and plasma in a mouse model of rTBIs [48].
Both t-tau and p-tau concentrations analyzed from plasma have thus far yielded variable results, with many studies failing to discern between people exposed to TBIs and controls [46][49][50][51][52][53]. Nevertheless, Alosco et al. [49] reported a relationship between tau levels and RHIs, as well as plasma t-tau levels above 3.56 pg/mL, only in former rugby players despite no significance between the groups. However, others reported no relationship between t-tau and RHI [52]. Only Olivera et al. [54] observed increased plasma t-tau levels in the military deployed within the last 18 months who self-diagnosed themselves with TBIs, with a greater number of TBIs associated with a more substantial increase in plasma t-tau. As for p-tau, Vasilevskaya et al. [53] showed that tau phosphorylated at threonine 181 (p-tau181) was significantly elevated in retired contact sports athletes.
While exosomal t-tau and p-tau appear promising as blood-based biomarkers, they so far have not shown substantial specificity to CTE; a significant increase in both plasma and exosomal t-tau and p-tau can be observed, for instance, in AD [55][56][57]. Studies on tau in CTE, thus far, have focused on its diagnostic utility in discerning between people with potential CTE and healthy controls, rather than between different neurodegenerative diseases. However, Turk et al. [22] reported that tau phosphorylated at threonine 231 (p-tau231) from CSF is significantly different between CTE and AD patients, confirmed by brain autopsies. Moreover, p-tau231 was successful in distinguishing AD and CTE diagnoses. There are no studies on the long-term blood levels of p-tau231, but the protein was significantly elevated in the plasma of patients from TBI rehabilitation units with a potential chronic TBI [58]

3.2. Amyloid Beta

While Amyloid beta (Aβ) plaques are primarily associated with AD, TBI has been shown to increase the concentration of the Amyloid Precursor Protein and Aβ peptides in the brain tissue and CSF. These proteins can foster the formation of plaques [9][59][60], which are toxic to brain cells and trigger neurodegenerative processes [61]. The presence of Aβ plaques in cadavers with neuropathologically-diagnosed CTE has been reported, but it is not as universal as the deposition of NFTs in CTE and has been linked to the possession of the Apoϵ4 allele, as well as significantly older age at death, potentially indicating Aβ plaques’ association with old age in CTE [9].
Thus far, the results for the blood-based Aβ peptide have been mixed. From plasma, Lebjman et al. [62] reported a significant increase in Aβ40 and a trend for increased Aβ42 in military personnel who experienced TBI, deployed a minimum of 16 months before the investigation, while other studies reported no significant changes in different groups of athletes who experienced TBIs [50][52]. Exosomal Aβ peptides, in turn, appear more promising, with a significant increase of Aβ42 in groups that experienced TBIs [63][64], with one study showing no difference [65]. While more research is needed to confirm the role of exosomal Aβ as a biomarker, its specificity to CTE is greatly limited.

3.3. Neurofilament Light

Neurofilaments are intermediate filaments expressed exclusively in neurons. While their exact function remains to be elucidated, they are thought to play a critical role in axonal stability. Therefore, the efflux of neurofilaments into the CSF and potentially systemic circulation is indicative of neuroaxonal injury and has been suggested as a biomarker of neurological disorders, such as Parkinson’s disease (PD) or AD [66]. In the context of TBI, the research has focused on the diagnosis of TBI through plasma NfL, reporting a significant elevation in plasma NfL following a TBI that predicted clinical outcomes [66][67][68][69][70]. However, knowledge about the long-term relationship between NfL and CTE is scarce. In a rat model, a single blast overpressure exposure was not shown to significantly increase plasma NfL 10 months after a blast simulation, but there was a trend for increased NfL in exposed rats compared to controls [71].
The major limitation of NfL is its lack of specificity to CTE. Asken et al. [72], analyzing a group of nine cadavers, showed that elevated NfL could be observed in patients with different neuropathologically confirmed neurological disorders, such as Frontotemporal Lobar Degeneration, CTE and AD. Nonetheless, further research into the chronic effects of TBIs and their relationship to NfL is warranted to establish the clinical relevance and selectivity of NfL in the context of CTE.

3.4. Other Biomarkers of Neurodegeneration

There have been individual studies investigating several other potential biomarkers of neurodegeneration. Ubiquitin C-Terminal Hydrolase L1 (UCH-L1) is an abundant protein in the brain and is essential to the proper maintenance of axonal integrity. Its dysfunction has been implicated in neurodegeneration, where it can, for example, misfold and constitute NFTs in AD [73]. As such, it was found to be significantly elevated in the CSF of AD patients [74][75]. In potential CTE, CSF UCH-L1 was associated with grey matter abnormalities in long-term TBI survivors, but there was no difference between this group and controls [46]. However, no UCH-L1 elevation, as well as no correlation with brain structural changes, was reported in patients with TBIs compared to controls, both from plasma [51] and exosomes [64], thus far.

4. Biomarkers of Neuroinflammation in CTE

4.1. Glial Fibrillary Acidic Protein

GFAP is an intermediate filament protein and a major cytoskeletal component of astrocytes, which maintain synaptic transmission and axonal metabolism [76]. Following TBI, astrocytes mediate processes, such as BBB permeability and the inflammatory response [77]. Astrocyte immune activation is accompanied by an increase in the expression of GFAP [78]. On the other hand, an astrocytic injury could cause the efflux of GFAP [79]. Therefore, GFAP could represent both chronic neuroinflammation and neurodegeneration in CTE. Its blood elevation has been shown to relate to structural abnormalities in imaging studies after mild TBI [80][81][82].

4.2. Inflammatory Cytokines

The increased activation of microglia, which has been shown to occur following TBIs, upregulates the production of several pro-inflammatory cytokines. These lead to increased permeability of the BBB, elevated secretion of chemokines that cause the migration of peripheral leukocytes into the brain, as well as the production of reactive oxygen species, which altogether foster neuroinflammation and can trigger secondary cell death. Inflammatory cytokines investigated in the context of the long-term consequences of TBI involve IL-6, IL-10, and TNF-α, which can all be secreted by microglia, indicating microgliosis [83]. As these can be expressed in all the tissues, the concentration of the cytokines was quantified from neuron-enriched exosomes.

5. Micro RNA Biomarkers in CTE

Micro RNAs (miRNAs) are small non-coding RNAs, which regulate a variety of processes at the post-transcriptional level. The expression of miRNAs can change in response to different physiological and pathological states [84]. Specifically, several studies identified panels of miRNA biomarkers from saliva [85] and blood [86][87][88][89] that showed different levels of specific miRNAs between patients following a TBI and controls. As such, they showed great potential in diagnosing TBI. A great advantage of miRNAs over conventional protein panels is their stability; Gilad et al. [84] showed that their levels did not change after four hours at room temperature, while two freeze–thaw cycles affected their levels to a small extent.
However, only a handful of studies have looked at the expression of miRNAs in potential CTE patients, thus far. Alvia et al. [90] compared the expression of different miRNAs previously indicated in the prefrontal cortex of brains donated by people who suffered from either CTE, Amyotrophic Lateral Sclerosis (ALS), or both. While much of the expression of miRNAs overlapped between CTE and ALS, they identified several miRNAs specific to CTE, which were involved in cell growth, apoptotic and inflammatory pathways. As per biological fluids, Ghai et al. [91] used next-generation sequencing (NGS) to compare the miRNA profiles of plasma and extracellular vesicles (EV) between veterans with a history of chronic TBI and controls. They detected significant differences in the levels of multiple previously described, as well as novel, miRNAs, which they confirmed using qRT-PCR. They also observed that most miRNAs were circulating freely in plasma, which supports the use of plasma without the need for EVs isolation. Ge et al. [92] compared serum and exosome biomarkers between 12 patients with a history of rTBIs and, thus, a different likelihood of CTE and respective controls, distinguishing serum and exosomal miR-1183 and exosomal miR-297 as potential diagnostic miRNA biomarkers of CTE. 

6. Conclusions

There are currently no approved biomarkers to diagnose CTE, thus it can only be confirmed through a post-mortem brain autopsy. Several imaging and cerebrospinal fluid biomarkers have shown promise in the diagnosis. However, blood-based biomarkers can be more easily obtained and quantified, increasing their clinical feasibility and potential for prophylactic use. This entry comprehensively summarizes the studies into potential blood-based biomarkers of CTE, discussing common themes and limitations, as well as suggesting future research directions. several molecules, such as different phosphorylated tau isoforms, were able to discern CTE from different neurodegenerative diseases. Further, the results from studies on exosomal biomarkers suggest that exosomes are a promising source of biomarkers, reflective of the internal environment of the brain. Nonetheless, more longitudinal studies combining imaging, neurobehavioral, and biochemical approaches are warranted to establish robust biomarkers for CTE.

References

  1. World Health Organization. Global Status Report on the Public Health Response to Dementia; WHO: Geneva, Switzerland, 2021.
  2. Martland, H.S. Punch Drunk. JAMA 1928, 91, 1103–1107.
  3. McKee, A.C.; Stein, T.D.; Nowinski, C.J.; Stern, R.A.; Daneshvar, D.H.; Alvarez, V.E.; Lee, H.-S.; Hall, G.; Wojtowicz, S.M.; Baugh, C.M.; et al. The spectrum of disease in chronic traumatic encephalopathy. Brain 2013, 136, 43–64.
  4. Arvanitakis, Z.; Shah, R.C.; Bennett, D.A. Diagnosis and Management of Dementia: Review. JAMA 2019, 322, 1589–1599.
  5. Robinson, L.; Tang, E.; Taylor, J.-P. Dementia: Timely diagnosis and early intervention. BMJ 2015, 350, h3029.
  6. Schwab, N.; Wennberg, R.; Grenier, K.; Tartaglia, C.; Tator, C.; Hazrati, L.-N. Association of Position Played and Career Duration and Chronic Traumatic Encephalopathy at Autopsy in Elite Football and Hockey Players. Neurology 2021, 96, e1835–e1843.
  7. Omalu, B.I.; DeKosky, S.T.; Minster, R.L.; Kamboh, M.I.; Hamilton, R.L.; Wecht, C.H. Chronic Traumatic Encephalopathy in a National Football League Player. Neurosurgery 2005, 57, 128–134.
  8. Omalu, B.I.; DeKosky, S.T.; Hamilton, R.L.; Minster, R.L.; Kamboh, M.I.; Shakir, A.M.; Wecht, C.H. Chronic Traumatic Encephalopathy in a National Football League Player: Part II. Neurosurgery 2006, 59, 1086–1093.
  9. Stein, T.D.; Montenigro, P.H.; Alvarez, V.E.; Xia, W.; Crary, J.F.; Tripodis, Y.; Daneshvar, D.H.; Mez, J.; Solomon, T.; Meng, G.; et al. Beta-amyloid deposition in chronic traumatic encephalopathy. Acta Neuropathol. 2015, 130, 21–34.
  10. Mez, J.; Daneshvar, D.H.; Kiernan, P.T.; Abdolmohammadi, B.; Alvarez, V.E.; Huber, B.R.; Alosco, M.L.; Solomon, T.M.; Nowinski, C.J.; McHale, L.; et al. Clinicopathological Evaluation of Chronic Traumatic Encephalopathy in Players of American Football. JAMA 2017, 318, 360–370.
  11. Goldstein, L.E.; Fisher, A.M.; Tagge, C.A.; Zhang, X.-L.; Velisek, L.; Sullivan, J.A.; Upreti, C.; Kracht, J.M.; Ericsson, M.; Wojnarowicz, M.W.; et al. Chronic Traumatic Encephalopathy in Blast-Exposed Military Veterans and a Blast Neurotrauma Mouse Model. Sci. Transl. Med. 2012, 4, 134.
  12. Maroon, J.C.; 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, e0117338.
  13. McKee, A.C.; Stein, T.D.; Huber, B.R.; Crary, J.F.; Bieniek, K.; Dickson, D.; Alvarez, V.E.; Cherry, J.D.; Farrell, K.; Butler, M.; et al. Chronic traumatic encephalopathy (CTE): Criteria for neuropathological diagnosis and relationship to repetitive head impacts. Acta Neuropathol. 2023, 145, 371–394.
  14. Stern, R.A.; Daneshvar, D.H.; Baugh, C.M.; Seichepine, D.R.; Montenigro, P.H.; Riley, D.O.; Fritts, N.G.; Stamm, J.M.; Robbins, C.A.; McHale, L.; et al. Clinical presentation of chronic traumatic encephalopathy. Neurology 2013, 81, 1122–1129.
  15. Gardner, R.C.; Yaffe, K. Epidemiology of mild traumatic brain injury and neurodegenerative disease. Mol. Cell. Neurosci. 2015, 66, 75–80.
  16. Guskiewicz, K.M.; Marshall, S.W.; Bailes, J.; McCrea, M.; Cantu, R.C.; Randolph, C.; Jordan, B.D. Association between Recurrent Concussion and Late-Life Cognitive Impairment in Retired Professional Football Players. Neurosurgery 2005, 57, 719–726.
  17. Perry, D.C.; Sturm, V.E.; Peterson, M.J.; Pieper, C.F.; Bullock, T.; Boeve, B.F.; Miller, B.L.; Guskiewicz, K.M.; Berger, M.S.; Kramer, J.H.; et al. Association of traumatic brain injury with subsequent neurological and psychiatric disease: A meta-analysis. JNS 2016, 124, 511–526.
  18. Barnes, D.E.; Byers, A.L.; Gardner, R.C.; Seal, K.H.; Boscardin, W.J.; Yaffe, K. Association of Mild Traumatic Brain Injury with and without Loss of Consciousness with Dementia in US Military Veterans. JAMA Neurol. 2018, 75, 1055–1061.
  19. Hind, K.; Konerth, N.; Entwistle, I.; Hume, P.; Theadom, A.; Lewis, G.; King, D.; Goodbourn, T.; Bottiglieri, M.; Ferraces-Riegas, P.; et al. Mental Health and Wellbeing of Retired Elite and Amateur Rugby Players and Non-contact Athletes and Associations with Sports-Related Concussion: The UK Rugby Health Project. Sports. Med. 2022, 52, 1419–1431.
  20. Alosco, M.L.; Culhane, J.; Mez, J. Neuroimaging Biomarkers of Chronic Traumatic Encephalopathy: Targets for the Academic Memory Disorders Clinic. Neurotherapeutics 2021, 18, 772–791.
  21. Alosco, M.L.; Tripodis, Y.; Fritts, N.G.; Heslegrave, A.; Baugh, C.M.; Conneely, S.; Mariani, M.; Martin, B.M.; Frank, S.; Mez, J.; et al. Cerebrospinal fluid tau, Aβ, and sTREM2 in Former National Football League Players: Modeling the relationship between repetitive head impacts, microglial activation, and neurodegeneration. Alzheimer’s Dement. 2018, 14, 1159–1170.
  22. Turk, K.W.; Geada, A.; Alvarez, V.E.; Xia, W.; Cherry, J.D.; Nicks, R.; Meng, G.; Daley, S.; Tripodis, Y.; Huber, B.R. A comparison between tau and amyloid-β cerebrospinal fluid biomarkers in chronic traumatic encephalopathy and Alzheimer disease. Alzheimer’s Res. Ther. 2022, 14, 28.
  23. Teunissen, C.E.; Verberk, I.M.W.; Thijssen, E.H.; Vermunt, L.; Hansson, O.; Zetterberg, H.; van der Flier, W.M.; Mielke, M.M.; del Campo, M. Blood-based biomarkers for Alzheimer’s disease: Towards clinical implementation. Lancet Neurol. 2022, 21, 66–77.
  24. Mckee, A.C.; Daneshvar, D.H. The neuropathology of traumatic brain injury. Handb. Clin. Neurol. 2015, 127, 45–66.
  25. Edwards, G.; Zhao, J.; Dash, P.K.; Soto, C.; Moreno-Gonzalez, I. Traumatic Brain Injury Induces Tau Aggregation and Spreading. J. Neurotrauma 2020, 37, 80–92.
  26. Butler, M.L.M.D.; Dixon, E.; Stein, T.D.; Alvarez, V.E.; Huber, B.; Buckland, M.E.; McKee, A.C.; Cherry, J.D. Tau Pathology in Chronic Traumatic Encephalopathy is Primarily Neuronal. J. Neuropathol. Exp. Neurol. 2022, 81, 773–780.
  27. Gendron, T.F.; Petrucelli, L. The role of tau in neurodegeneration. Mol. Neurodegener. 2009, 4, 13.
  28. McKee, A.C.; Cantu, R.C.; Nowinski, C.J.; Hedley-Whyte, E.T.; Gavett, B.E.; Budson, A.E.; Santini, V.E.; Lee, H.-S.; Kubilus, C.A.; Stern, R.A. Chronic Traumatic Encephalopathy in Athletes: Progressive Tauopathy After Repetitive Head Injury. J. Neuropathol. Exp. Neurol. 2009, 68, 709–735.
  29. McKee, A.C.; Stein, T.D.; Kiernan, P.T.; Alvarez, V.E. The Neuropathology of Chronic Traumatic Encephalopathy: CTE Neuropathology. Brain Pathol. 2015, 25, 350–364.
  30. Smith, D.H.; Meaney, D.F.; Shull, W.H. Diffuse Axonal Injury in Head Trauma. J. Head Trauma Rehabil. 2003, 18, 307–316.
  31. Inglese, M.; Makani, S.; Johnson, G.; Cohen, B.A.; Silver, J.A.; Gonen, O.; Grossman, R.I. Diffuse axonal injury in mild traumatic brain injury: A diffusion tensor imaging study. J. Neurosurg. 2005, 103, 298–303.
  32. Petzold, A. Neurofilament phosphoforms: Surrogate markers for axonal injury, degeneration and loss. J. Neurol. Sci. 2005, 233, 183–198.
  33. Siedler, D.G.; Chuah, M.I.; Kirkcaldie, M.T.K.; Vickers, J.C.; King, A.E. Diffuse axonal injury in brain trauma: Insights from alterations in neurofilaments. Front. Cell. Neurosci. 2014, 8, 429.
  34. Hay, J.R.; Johnson, V.E.; Young, A.M.H.; Smith, D.H.; Stewart, W. Blood-Brain Barrier Disruption Is an Early Event That May Persist for Many Years After Traumatic Brain Injury in Humans. J. Neuropathol. Exp. Neurol. 2015, 74, 1147–1157.
  35. Kornguth, S.; Rutledge, N.; Perlaza, G.; Bray, J.; Hardin, A. A Proposed Mechanism for Development of CTE Following Concussive Events: Head Impact, Water Hammer Injury, Neurofilament Release, and Autoimmune Processes. Brain Sci. 2017, 7, 164.
  36. Mayeux, R. Biomarkers: Potential uses and limitations. Neurotherapeutics 2004, 1, 182–188.
  37. Ray, P.; Manach, Y.L.; Riou, B.; Houle, T.T.; Warner, D.S. Statistical Evaluation of a Biomarker. Anesthesiology 2010, 112, 1023–1040.
  38. Shahim, P.; Gill, J.M.; Blennow, K.; Zetterberg, H. Fluid Biomarkers for Chronic Traumatic Encephalopathy. Semin. Neurol. 2020, 40, 411–419.
  39. Johnson, V.E.; Stewart, J.E.; Begbie, F.D.; Trojanowski, J.Q.; Smith, D.H.; Stewart, W. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain 2013, 136, 28–42.
  40. Erturk, A.; Mentz, S.; Stout, E.E.; Hedehus, M.; Dominguez, S.L.; Neumaier, L.; Krammer, F.; Llovera, G.; Srinivasan, K.; Hansen, D.V. Interfering with the Chronic Immune Response Rescues Chronic Degeneration After Traumatic Brain Injury. J. Neurosci. 2016, 36, 9962–9975.
  41. McKee, A.C. The Neuropathology of Chronic Traumatic Encephalopathy: The Status of the Literature. Semin. Neurol. 2020, 40, 359–369.
  42. Murray, H.C.; Osterman, C.; Bell, P.; Vinnell, L.; Curtis, M.A. Neuropathology in chronic traumatic encephalopathy: A systematic review of comparative post-mortem histology literature. Acta Neuropathol. Commun. 2022, 10, 108–128.
  43. Johnson, V.E.; Stewart, W.; Smith, D.H. Widespread Tau and Amyloid-Beta Pathology Many Years After a Single Traumatic Brain Injury in Humans: Long-Term AD-Like Pathology after Single TBI. Brain Pathol. 2012, 22, 142–149.
  44. Alosco, M.L.; Su, Y.; Stein, T.D.; Protas, H.; Cherry, J.D.; Adler, C.H.; Balcer, L.J.; Bernick, C.; Pulukuri, S.V.; Abdolmohammadi, B.; et al. Associations between near end-of-life flortaucipir PET and postmortem CTE-related tau neuropathology in six former American football players. Eur. J Nucl. Med. Mol. Imaging 2022, 50, 435–452.
  45. Mohamed, A.Z.; Cumming, P.; Nasrallah, F.A.; Alzheimer’s Disease Neuroimaging Initiative. Escalation of Tau Accumulation after a Traumatic Brain Injury: Findings from Positron Emission Tomography. Brain Sci. 2022, 12, 876.
  46. Gorgoraptis, N.; Li, L.M.; Whittington, A.; Zimmerman, K.A.; Maclean, L.M.; McLeod, C.; Ross, E.; Heslegrave, A.; Zetterberg, H.; Passchier, J.; et al. In vivo detection of cerebral tau pathology in long-term survivors of traumatic brain injury. Sci. Transl. Med. 2019, 11, eaaw1993.
  47. Muraoka, S.; Jedrychowski, M.P.; Tatebe, H.; DeLeo, A.M.; Ikezu, S.; Tokuda, T.; Gygi, S.P.; Stern, R.A.; Ikezu, T. Proteomic Profiling of Extracellular Vesicles Isolated from Cerebrospinal Fluid of Former National Football League Players at Risk for Chronic Traumatic Encephalopathy. Front. Neurosci. 2019, 13, 1059.
  48. Rubenstein, R.; Sharma, D.R.; Chang, B.; Oumata, N.; Cam, M.; Vaucelle, L.; Lindberg, M.F.; Chiu, A.; Wisniewski, T.; Wang, K.K.W.; et al. Novel Mouse Tauopathy Model for Repetitive Mild Traumatic Brain Injury: Evaluation of Long-Term Effects on Cognition and Biomarker Levels After Therapeutic Inhibition of Tau Phosphorylation. Front. Neurol. 2019, 10, 124.
  49. Alosco, M.L.; Tripodis, Y.; Jarnagin, J.; Baugh, C.M.; Martin, B.; Chaisson, C.E.; Estochen, N.; Song, L.; Cantu, R.C.; Jeromin, A.; et al. Repetitive head impact exposure and later-life plasma total tau in former National Football League players. Alzheimer’s Dement. Diagn. Assess. Dis. Monit. 2017, 7, 33–40.
  50. Swann, O.J.; Turner, M.; Heslegrave, A.; Zetterberg, H. Fluid biomarkers and risk of neurodegenerative disease in retired athletes with multiple concussions: Results from the International Concussion and Head Injury Research Foundation Brain health in Retired athletes Study of Ageing and Impact-Related Neurodegenerative Disease (ICHIRF-BRAIN study). BMJ Open Sport Exerc. Med. 2022, 8, e001327.
  51. Shahim, P.; Politis, A.; van der Merwe, A.; Moore, B.; Ekanayake, V.; Lippa, S.M.; Chou, Y.-Y.; Pham, D.L.; Butman, J.A.; Diaz-Arrastia, R.; et al. Time course and diagnostic utility of NfL, tau, GFAP, and UCH-L1 in subacute and chronic TBI. Neurology 2020, 95, e623–e636.
  52. Shahim, P.; Zetterberg, H.; Simrén, J.; Ashton, N.J.; Norato, G.; Schöll, M.; Tegner, Y.; Diaz-Arrastia, R.; Blennow, K. Association of Plasma Biomarker Levels with Their CSF Concentration and the Number and Severity of Concussions in Professional Athletes. Neurology 2022, 99, e347–e354.
  53. Vasilevskaya, A.; Taghdiri, F.; Multani, N.; Ozzoude, M.; Tarazi, A.; Khodadadi, M.; Wennberg, R.; Rusjan, P.; Houle, S.; Green, R.; et al. Investigating the use of plasma pTau181 in retired contact sports athletes. J. Neurol. 2022, 269, 5582–5595.
  54. Olivera, A.; Lejbman, N.; Jeromin, A.; French, L.M.; Kim, H.-S.; Cashion, A.; Mysliwiec, V.; Diaz-Arrasti, R.; Gill, J. Peripheral Total Tau in Military Personnel Who Sustain Traumatic Brain Injuries During Deployment. JAMA Neurol. 2015, 72, 1109.
  55. Yin, Q.; Ji, X.; Lv, R.; Pei, J.-J.; Du, Y.; Shen, C.; Hou, X. Targetting Exosomes as a New Biomarker and Therapeutic Approach for Alzheimer’s Disease. CIA 2020, 15, 195–205.
  56. Asken, B.M.; Tanner, J.A.; VandeVrede, L.; Mantyh, W.G.; Casaletto, K.B.; Staffaroni, A.M.; La Joie, R.; Iaccarino, L.; Soleimani-Meigooni, D.; Rojas, J.C. Plasma P-tau181 and P-tau217 in Patients with Traumatic Encephalopathy Syndrome with and without Evidence of Alzheimer Disease Pathology. Neurology 2022, 99, e594–e604.
  57. Varesi, A.; Carrara, A.; Pires, V.G.; Floris, V.; Pierella, E.; Savioli, G.; Prasad, S.; Esposito, C.; Ricevuti, G.; Chirumbolo, S.; et al. Blood-Based Biomarkers for Alzheimer’s Disease Diagnosis and Progression: An Overview. Cells 2022, 11, 1367.
  58. Rubenstein, R.; Chang, B.; Yue, J.K.; Chiu, A.; Winkler, E.A.; Puccio, A.M.; Diaz-Arrastia, R.; Yuh, E.L.; Mukherjee, P.; Valadka, A.B.; et al. Comparing Plasma Phospho Tau, Total Tau, and Phospho Tau–Total Tau Ratio as Acute and Chronic Traumatic Brain Injury Biomarkers. JAMA Neurol. 2017, 74, 1063.
  59. Smith, D.H.; Chen, X.H.; Iwata, A.; Graham, D.I. Amyloid beta accumulation in axons after traumatic brain injury in humans. J. Neurosurg. 2003, 98, 1072–1077.
  60. Olsson, A.; Csajbok, L.; Ost, M.; Höglund, K.; Nylén, K.; Rosengren, L.; Nellgård, B.; Blennow, K. Marked increase of beta-amyloid(1–42) and amyloid precursor protein in ventricular cerebrospinal fluid after severe traumatic brain injury. J. Neurol. 2004, 251, 870–876.
  61. Gupta, A.; Goyal, R. Amyloid beta plaque: A culprit for neurodegeneration. Acta Neurol. Belg. 2016, 116, 445–450.
  62. Lejbman, N.; Olivera, A.; Heinzelmann, M.; Feng, R.; Yun, S.; Kim, H.-S.; Gill, J. Active duty service members who sustain a traumatic brain injury have chronically elevated peripheral concentrations of A β 40 and lower ratios of A β 42/40. Brain Inj. 2016, 30, 1436–1441.
  63. Gill, J.; Mustapic, M.; Diaz-Arrastia, R.; Lange, R.; Gulyani, S.; Diehl, T.; Motamedi, V.; Osier, N.; Stern, R.A.; Kapogiannis, D. Higher exosomal tau, amyloid-beta 42 and IL-10 are associated with mild TBIs and chronic symptoms in military personnel. Brain Inj. 2018, 32, 1359–1366.
  64. Goetzl, E.J.; Elahi, F.M.; Mustapic, M.; Kapogiannis, D.; Pryhoda, M.; Gilmore, A.; Gorgens, K.A.; Davidson, B.; Granholm, A.; Ledreux, A. Altered levels of plasma neuron-derived exosomes and their cargo proteins characterize acute and chronic mild traumatic brain injury. FASEB J. 2019, 33, 5082–5088.
  65. Peltz, C.B.; Kenney, K.; Gill, J.; Diaz-Arrastia, R.; Gardner, R.C.; Yaffe, K. Blood biomarkers of traumatic brain injury and cognitive impairment in older veterans. Neurology 2020, 95, e1126–e1133.
  66. Khalil, M.; Teunissen, C.E.; Otto, M.; Piehl, F.; Sormani, M.P.; Gattringer, T.; Barro, C.; Kappos, L.; Comabella, M.; Fazekas, F.; et al. Neurofilaments as biomarkers in neurological disorders. Nat. Rev. Neurol. 2008, 14, 577–589.
  67. Al Nimer, F.; Thelin, E.; Nyström, H.; Dring, A.M.; Svenningsson, A.; Piehl, F.; Nelson, D.W.; Bellander, B.M. Comparative Assessment of the Prognostic Value of Biomarkers in Traumatic Brain Injury Reveals an Independent Role for Serum Levels of Neurofilament Light. PLoS ONE 2015, 10, e0132177.
  68. Shahim, P.; Gren, M.; Liman, V.; Andreasson, U.; Norgren, N.; Tegner, Y.; Mattsson, N.; Andreasen, N.; Ost, M.; Zetterberg, H.; et al. Serum neurofilament light protein predicts clinical outcome in traumatic brain injury. Sci. Rep. 2016, 6, 36791.
  69. Boutté, A.M.; Thangavelu, B.; LaValle, C.R.; Nemes, J.; Gilsdorf, J.; Shear, D.A.; Kamimori, G.H. Brain-related proteins as serum biomarkers of acute, subconcussive blast overpressure exposure: A cohort study of military personnel. PLoS ONE 2019, 14, e0221036.
  70. Gao, W.; Zhang, Z.; Lv, X.; Wu, Q.; Yan, J.; Mao, G.; Xing, W. Neurofilament light chain level in traumatic brain injury: A system review and meta-analysis. Medicine 2020, 99, e22363.
  71. Dickstein, D.L.; De Gasperi, R.; Gama Sosa, M.A.; Perez-Garcia, G.; Short, J.A.; Sosa, H.; Perez, G.M.; Tschiffely, A.E.; Dams-O’Connor, K.; Pullman, M.Y.; et al. Brain and blood biomarkers of tauopathy and neuronal injury in humans and rats with neurobehavioral syndromes following blast exposure. Mol. Psychiatry 2021, 26, 5940–5954.
  72. Asken, B.M.; Tanner, J.A.; Vande-Vrede, L.; Casaletto, K.B.; Staffaroni, A.M.; Mundada, N.; Fonseca, C.; Iaccarino, L.; La Joie, R.; Tsuei, T.; et al. Multi-Modal Biomarkers of Repetitive Head Impacts and Traumatic Encephalopathy Syndrome: A Clinicopathological Case Series. J. Neurotrauma 2022, 39, 1195–1213.
  73. Bishop, P.; Rocca, D.; Henley, J.M. Ubiquitin C-terminal hydrolase L1 (UCH-L1): Structure, distribution and roles in brain function and dysfunction. Biochem. J. 2016, 473, 2453–2462.
  74. Öhrfelt, A.; Johansson, P.; Wallin, A.; Andreasson, U.; Zetterberg, H.; Blennow, K.; Svensson, J. Increased Cerebrospinal Fluid Levels of Ubiquitin Carboxyl-Terminal Hydrolase L1 in Patients with Alzheimer’s Disease. Dement. Geriatr. Cogn. Disord. Extra 2016, 6, 283–294.
  75. Kulczynska-Przybik, A.; Dulewicz, M.; Mroczko, P.; Borawska, R.; Doroszkiewicz, J.; Litman-Zawadzka, A.; Arslan, D.; Slowik, A. The assessment of ubiquitin C-terminal hydrolase-1 (UCH-L1) in patients with Alzheimer’s disease. Alzheimer’s Dement. 2022, 18, e062156.
  76. Sofroniew, M.V.; Vinters, H.V. Astrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 7–35.
  77. Burda, J.E.; Bernstein, A.M.; Sofroniew, M.V. Astrocyte roles in traumatic brain injury. Exp. Neurol. 2016, 275, 305–315.
  78. Liddelow, S.A.; Barres, B.A. Reactive astrocytes: Production, function, and therapeutic potential. Immunity 2017, 46, 957–967.
  79. Abdelhak, A.; Foschi, M.; Abu-Rumeileh, S.; Yue, J.K.; D’Anna, L.; Huss, A.; Oeckl, P.; Ludolph, A.C.; Kuhle, J.; Petzold, A. Blood GFAP as an emerging biomarker in brain and spinal cord disorders. Nat. Rev. Neurol. 2022, 18, 158–172.
  80. Lumpkins, K.M.; Bochicchio, G.V.; Keledjian, K.; Simard, J.M.; McCunn, M.; Scalea, T. Glial fibrillary acidic protein is highly correlated with brain injury. J. Trauma 2008, 65, 778–784.
  81. Gill, J.; Latour, L.; Diaz-Arrastia, R.; Motamedi, V.; Turtzo, C.; Shahim, P.; Mondello, S.; DeVoto, C.; Veras, E.; Hanlon, D.; et al. Glial fibrillary acidic protein elevations relate to neuroimaging abnormalities after mild TBI. Neurology 2018, 91, e1385–e1389.
  82. Huebschmann, N.A.; Luoto, T.M.; Karr, J.E.; Berghem, K.; Blennow, K.; Zetterberg, H.; Ashton, N.J.; Simrén, J.; Posti, J.P.; Gill, J.M.; et al. Comparing Glial Fibrillary Acidic Protein (GFAP) in Serum and Plasma Following Mild Traumatic Brain Injury in Older Adults. Front. Neurol. 2020, 11, 1054.
  83. Lozano, D.; Gonzales-Portillo, G.S.; Acosta, S.; de la Pena, I.; Tajiri, N.; Kaneko, Y.; Borlongan, C.V. Neuroinflammatory responses to traumatic brain injury: Etiology, clinical consequences, and therapeutic opportunities. Neuropsychiatr. Dis. Treat. 2015, 11, 97–106.
  84. Gilad, S.; Meiri, E.; Yogev, Y.; Benjamin, S.; Lebanony, D.; Yerushalmi, N.; Benjamin, H.; Kushnir, M.; Cholakh, H.; Melamed, N.; et al. Serum MicroRNAs Are Promising Novel Biomarkers. PLoS ONE 2008, 3, e3148.
  85. Hiskens, M.I.; Mengistu, T.S.; Li, K.M.; Fenning, A.S. Systematic Review of the Diagnostic and Clinical Utility of Salivary microRNAs in Traumatic Brain Injury (TBI). Int. J. Mol. Sci. 2022, 23, 13160.
  86. Guedes, V.A.; Devoto, C.; Leete, J.; Sass, D.; Acott, J.D.; Mithani, S.; Gill, J.M. Extracellular Vesicle Proteins and MicroRNAs as Biomarkers for Traumatic Brain Injury. Front. Neurol. 2020, 11, 663.
  87. Bhomia, M.; Balakathiresan, N.S.; Wang, K.K.; Papa, L.; Maheshwari, R.K. A Panel of Serum MiRNA Biomarkers for the Diagnosis of Severe to Mild Traumatic Brain Injury in Humans. Sci. Rep. 2016, 6, 28148.
  88. Redell, J.B.; Moore, A.N.; Ward, N.H.; Hergenroeder, G.W.; Dash, P.K. Human Traumatic Brain Injury Alters Plasma microRNA Levels. J. Neurotrauma 2010, 27, 2147–2156.
  89. Wyczechowska, D.; Harch, P.G.; Mullenix, S.; Fannin, E.S.; Chiappinelli, B.B.; Jeansonne, D.; Lassak, A.; Bazan, N.G.; Peruzzi, F. Serum microRNAs associated with concussion in football players. Front. Neurol. 2023, 14, 1155479.
  90. Alvia, M.; Aytan, N.; Spencer, K.R.; Foster, Z.W.; Rauf, N.A.; Guilderson, L.; Robey, I.; Averill, J.G.; Walker, S.E.; Alvarez, V.E.; et al. MicroRNA Alterations in Chronic Traumatic Encephalopathy and Amyotrophic Lateral Sclerosis. Front. Neurosci. 2022, 16, 855096.
  91. Ghai, V.; Fallen, S.; Baxter, D.; Scherler, K.; Kim, T.-K.; Zhou, Y.; Meabon, J.S.; Logsdon, A.F.; Banks, W.A.; Schindler, A.G.; et al. Alterations in Plasma microRNA and Protein Levels in War Veterans with Chronic Mild Traumatic Brain Injury. J. Neurotrauma 2020, 37, 1418–1430.
  92. Ge, X.; Guo, M.; Li, M.; Zhang, S.; Qiang, J.; Zhu, L.; Cheng, L.; Li, W.; Wang, Y.; Yu, J.; et al. Potential blood biomarkers for chronic traumatic encephalopathy: The multi-omics landscape of an observational cohort. Front. Aging Neurosci. 2022, 14, 1052765.
More
Information
Subjects: Clinical Neurology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 812
Revisions: 2 times (View History)
Update Date: 29 Aug 2023
1000/1000