Submitted:
24 October 2023
Posted:
25 October 2023
You are already at the latest version
Abstract
Keywords:
1. Background
2. Neuropathology
3. Potential TES Biomarkers
3.1. Neuroimaging
3.2. CSF
3.3. Plasma
4. Treatment
4.1. Non-Pharmacological Management
4.2. Clinical Pharmacological Management
4.3. Clinical Use Of Nutraceutical Regimen
4.4. Preclinical Investigational Pharmacological Intervention
4.5. Targeting Tau Acetylation
4.6. Targeting Tau Phosphorylation
4.7. Targeting Inflammation
4.8. Immunotherapy
4.9. Potential dietary targets
5. Future Directions
| TBI Pharmacological Regimen | Proposed Mechanism |
|---|---|
| Cholinesterase Inhibitors | Cholinesterase inhibitors including galantamine, donepezil, and rivastigmine, have been repurposed for TBI patients87. |
| NMDA receptor antagonists | NMDA receptor antagonist, amantadine, has been shown to improve cognition in moderate to severe TBI patients88. |
| SSRIs | Selective serotonin reuptake inhibitors (SSRIs) like sertraline and escitalopram have been utilized to mange behavioral symptoms in TBI patients89. |
| Guanfacine | Guanfacine has been reported to improve working memory deficits in mild TBI patients90. |
| Nutraceuticals | A number of nutraceuticals have been utilized in treatment of TBI preclinical and clinical studies, including N-acetylcysteine (NAC), flavonoids, resveratrol, alpha-tocopherol (vitamin E), coenzyme Q1091. |
| NSAIDs | COX-2 selective drugs like carprofen, celecoxib, meloxicam, nimesulide, and rofecoxib have undergone testing in various preclinical TBI models, with no significant degree of established efficacy123. |
| Glucocorticoids | Despite several promising preclinical studies, clinical trials have resulted in limited success, likely due to a narrow therapeutic window137. |
| Phosphodiesterase Inhibitors | Phophodiesterase inhibitors have been utilized mostly in preclinical studies, and have not systematically studied in clinical trial setting146. |
| Minocycline | In prior preclinical studies, minocycline given between 5 min and 1 h after injury improved performance on a variety of neurobehavioral150. |
| Progesterone | A large, multi-center Phase III PROtect III trial, , as well as a second larger scale trial (SYNAPSE) examined progesterone did not establish clinical effiacy156. |
| Erythropoietin | Despite preclinical studies success, the evidence for the use of erythropoietin has not reached the threshold for its use in a phase III trial123. |
| Anakinra | A small phase II randomized controlled clinical trial reported anti-inflammatory benefits in Anakinra treated group, but the study size was too small to establish efficacy, but provided an intriguing potential future approach144. |
| Tau phosphorylation targets | The studies focusing on tau-phosphorylation targets have been mostly preclinical, with possible future clinical applications94. |
| Tau acetylation targets | Tau acetylation inhibitors including salsalate, as well as methylene blue, as well as histone deacetylase 6 and sirtuins have largely been examined in the preclinical setting109. |
| Immunotherapy | Specific antibodies targeting the pathogenic cis-P-tau post TBI have been reported to lead to improved structural and functional outcomes165, but yet to be examine in larger clinical trial setting. |
References
- Asken, B.M. et al. Alzheimer’s pathology is associated with altered cognition, brain volume, and plasma biomarker patterns in traumatic encephalopathy syndrome. Alzheimer's Research & Therapy 15, 126 (2023). [CrossRef]
- McKee, A.C. et al. The first NINDS/NIBIB consensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol 131, 75-86 (2016). [CrossRef]
- Martland, H.S. Punch drunk. Journal of the American Medical Association 91, 1103-1107 (1928). [CrossRef]
- Castellani, R.J. & Perry, G. Dementia pugilistica revisited. Journal of Alzheimer's disease 60, 1209-1221 (2017). [CrossRef]
- Asken, B.M., Sullan, M.J., DeKosky, S.T., Jaffee, M.S. & Bauer, R.M. Research gaps and controversies in chronic traumatic encephalopathy: A review. JAMA neurology 74, 1255-1262 (2017). [CrossRef]
- Omalu, B.I., Bailes, J., Hammers, J.L. & Fitzsimmons, R.P. Chronic traumatic encephalopathy, suicides and parasuicides in professional American athletes: The role of the forensic pathologist. The American journal of forensic medicine and pathology 31, 130-132 (2010). [CrossRef]
- Omalu, B.I. et al. Chronic traumatic encephalopathy in a national football league player: Part II. Neurosurgery 59, 1086-1092; discussion 1092-1083 (2006). [CrossRef]
- Omalu, B.I. et al. Chronic traumatic encephalopathy in a National Football League player. Neurosurgery 57, 128-134; discussion 128-134 (2005). [CrossRef]
- Montenigro, P.H. et al. Clinical subtypes of chronic traumatic encephalopathy: Literature review and proposed research diagnostic criteria for traumatic encephalopathy syndrome. Alzheimer's research & therapy 6, 1-17 (2014). [CrossRef]
- Mez, J. et al. Validity of the 2014 traumatic encephalopathy syndrome criteria for CTE pathology. Alzheimer's & Dementia 17, 1709-1724 (2021). [CrossRef]
- Kelly, J.P., Priemer, D.S., Perl, D.P. & Filley, C.M. Sports concussion and chronic traumatic encephalopathy: Finding a path forward. Annals of neurology 93, 222-225 (2023). [CrossRef]
- Corsellis, J.A.N., Bruton, C.J. & Freeman-Browne, D. The aftermath of boxing. Psychological Medicine 3, 270-303 (1973). [CrossRef]
- McKee, A.C. et al. The spectrum of disease in chronic traumatic encephalopathy. Brain 136, 43-64 (2013). [CrossRef]
- Alosco, M.L. et al. Developing methods to detect and diagnose chronic traumatic encephalopathy during life: Rationale, design, and methodology for the DIAGNOSE CTE Research Project. Alzheimer's research & therapy 13, 1-23 (2021). [CrossRef]
- Falcon, B. et al. Novel tau filament fold in chronic traumatic encephalopathy encloses hydrophobic molecules. Nature 568, 420-423 (2019). [CrossRef]
- McKee, A.C., Stein, T.D., Kiernan, P.T. & Alvarez, V.E. The neuropathology of chronic traumatic encephalopathy. Brain pathology 25, 350-364 (2015). [CrossRef]
- Gardner, A., Iverson, G.L. & McCrory, P. Chronic traumatic encephalopathy in sport: A systematic review. British journal of sports medicine 48, 84-90 (2014). [CrossRef]
- Bieniek, K.F. et al. Association between contact sports participation and chronic traumatic encephalopathy: A retrospective cohort study. Brain Pathology 30, 63-74 (2020). [CrossRef]
- Stern, R.A. et al. Clinical presentation of chronic traumatic encephalopathy. Neurology 81, 1122-1129 (2013). [CrossRef]
- Hampshire, A., MacDonald, A. & Owen, A.M. Hypoconnectivity and hyperfrontality in retired American football players. Scientific reports 3, 1-8 (2013). [CrossRef]
- Asken, B.M. et al. Multi-Modal Biomarkers of Repetitive Head Impacts and Traumatic Encephalopathy Syndrome: A Clinicopathological Case Series. Journal of Neurotrauma 39, 1195-1213 (2022). [CrossRef]
- Strain, J. et al. Depressive symptoms and white matter dysfunction in retired NFL players with concussion history. Neurology 81, 25-32 (2013). [CrossRef]
- Turk, K.W. & Budson, A.E. Chronic traumatic encephalopathy. CONTINUUM: Lifelong Learning in Neurology 25, 187-207 (2019). [CrossRef]
- Stern, R.A. et al. Tau positron-emission tomography in former national football league players. New England journal of medicine 380, 1716-1725 (2019). [CrossRef]
- Grashow, R. et al. Premortem chronic traumatic encephalopathy diagnoses in professional football. Annals of neurology 88, 106-112 (2020). [CrossRef]
- Alosco, M.L. 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 & Dementia 14, 1159-1170 (2018). [CrossRef]
- Asken, B.M. et al. Plasma glial fibrillary acidic protein levels differ along the spectra of amyloid burden and clinical disease stage. Journal of Alzheimer's Disease 78, 265-276 (2020). [CrossRef]
- Benedet, A.L. et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA neurology 78, 1471-1483 (2021). [CrossRef]
- Pereira, J.B. et al. Plasma GFAP is an early marker of amyloid-β but not tau pathology in Alzheimer’s disease. Brain 144, 3505-3516 (2021). [CrossRef]
- Wang, K.K. et al. An update on diagnostic and prognostic biomarkers for traumatic brain injury. Expert review of molecular diagnostics 18, 165-180 (2018). [CrossRef]
- Böhmer, A.E. et al. Neuron-specific enolase, S100B, and glial fibrillary acidic protein levels as outcome predictors in patients with severe traumatic brain injury. Neurosurgery 68, 1624-1631 (2011). [CrossRef]
- Stein, D.M. et al. Association of CSF biomarkers and secondary insults following severe traumatic brain injury. Neurocritical care 14, 200-207 (2011). [CrossRef]
- Berger, R.P. et al. Neuron-specific enolase and S100B in cerebrospinal fluid after severe traumatic brain injury in infants and children. Pediatrics 109, e31-e31 (2002). [CrossRef]
- Liu, M. et al. A novel rat model of blast-induced traumatic brain injury simulating different damage degree: Implications for morphological, neurological, and biomarker changes. Frontiers in cellular neuroscience 9, 168 (2015). [CrossRef]
- Topolovec-Vranic, J. et al. The value of serum biomarkers in prediction models of outcome after mild traumatic brain injury. Journal of Trauma and Acute Care Surgery 71, S478-S486 (2011). [CrossRef]
- Buonora, J.E. et al. Multivariate analysis of traumatic brain injury: Development of an assessment score. Frontiers in neurology 6, 68 (2015). [CrossRef]
- Verfaillie, C.J. & Delanghe, J.R. Hemolysis correction factor in the measurement of serum neuron-specific enolase. Clinical chemistry and laboratory medicine 48, 891-892 (2010). [CrossRef]
- Kobeissy, F.H. et al. Novel differential neuroproteomics analysis of traumatic brain injury in rats. Molecular & Cellular Proteomics 5, 1887-1898 (2006). [CrossRef]
- Mondello, S. et al. Clinical utility of serum levels of ubiquitin C-terminal hydrolase as a biomarker for severe traumatic brain injury. Neurosurgery 70, 666 (2012). [CrossRef]
- Brophy, G.M. et al. Biokinetic analysis of ubiquitin C-terminal hydrolase-L1 (UCH-L1) in severe traumatic brain injury patient biofluids. Journal of neurotrauma 28, 861-870 (2011). [CrossRef]
- Blyth, B.J. et al. Validation of serum markers for blood-brain barrier disruption in traumatic brain injury. Journal of neurotrauma 26, 1497-1507 (2009). [CrossRef]
- Papa, L. et al. Ubiquitin C-terminal hydrolase is a novel biomarker in humans for severe traumatic brain injury. Critical care medicine 38, 138 (2010). [CrossRef]
- Zhang, Z.-Y. et al. Comparison of the performances of copeptin and multiple biomarkers in long-term prognosis of severe traumatic brain injury. Peptides 60, 13-17 (2014). [CrossRef]
- Diaz-Arrastia, R. et al. Acute biomarkers of traumatic brain injury: Relationship between plasma levels of ubiquitin C-terminal hydrolase-L1 and glial fibrillary acidic protein. Journal of neurotrauma 31, 19-25 (2014). [CrossRef]
- Papa, L. et al. Elevated levels of serum glial fibrillary acidic protein breakdown products in mild and moderate traumatic brain injury are associated with intracranial lesions and neurosurgical intervention. Annals of emergency medicine 59, 471-483 (2012). [CrossRef]
- Bazarian, J.J. et al. Serum GFAP and UCH-L1 for prediction of absence of intracranial injuries on head CT (ALERT-TBI): A multicentre observational study. The Lancet Neurology 17, 782-789 (2018). [CrossRef]
- Korley, F.K. et al. Comparison of GFAP and UCH-L1 measurements from two prototype assays: The Abbott i-STAT and ARCHITECT assays. Neurotrauma reports 2, 193-199 (2021). [CrossRef]
- Schulte, S., Podlog, L.W., Hamson-Utley, J.J., Strathmann, F.G. & Strüder, H.K. A systematic review of the biomarker S100B: Implications for sport-related concussion management. Journal of Athletic Training 49, 830-850 (2014). [CrossRef]
- Filippidis, A.S., Papadopoulos, D.C., Kapsalaki, E.Z. & Fountas, K.N. Role of the S100B serum biomarker in the treatment of children suffering from mild traumatic brain injury. Neurosurgical Focus 29, E2 (2010). [CrossRef]
- Kiechle, K. et al. Subject-specific increases in serum S-100B distinguish sports-related concussion from sports-related exertion. PloS one 9, e84977 (2014). [CrossRef]
- Cervellin, G. et al. Serum levels of protein S100B predict intracranial lesions in mild head injury. Clinical biochemistry 45, 408-411 (2012). [CrossRef]
- Papa, L. et al. GFAP out-performs S100β in detecting traumatic intracranial lesions on computed tomography in trauma patients with mild traumatic brain injury and those with extracranial lesions. Journal of neurotrauma 31, 1815-1822 (2014). [CrossRef]
- Metting, Z., Wilczak, N., Rodiger, L., Schaaf, J. & Van Der Naalt, J. GFAP and S100B in the acute phase of mild traumatic brain injury. Neurology 78, 1428-1433 (2012). [CrossRef]
- Barbosa, R.R. et al. Evaluation and management of mild traumatic brain injury: An Eastern Association for the Surgery of Trauma practice management guideline. Journal of Trauma and Acute Care Surgery 73, S307-S314 (2012). [CrossRef]
- Zongo, D. et al. S100-B protein as a screening tool for the early assessment of minor head injury. Annals of emergency medicine 59, 209-218 (2012). [CrossRef]
- Pike, B.R. et al. Accumulation of calpain and caspase-3 proteolytic fragments of brain-derived αII-spectrin in cerebral spinal fluid after middle cerebral artery occlusion in rats. Journal of Cerebral Blood Flow & Metabolism 24, 98-106 (2004). [CrossRef]
- Mondello, S. et al. αII-spectrin breakdown products (SBDPs): Diagnosis and outcome in severe traumatic brain injury patients. Journal of neurotrauma 27, 1203-1213 (2010). [CrossRef]
- Vartanian, M.G. et al. Phenytoin pretreatment prevents hypoxic-ischemic brain damage in neonatal rats. Developmental brain research 95, 169-175 (1996). [CrossRef]
- Berger, R.P. et al. Translating biomarkers research to clinical care: Applications and issues for rehabilomics. PM&R 3, S31-S38 (2011). [CrossRef]
- Pike, B.R. et al. Accumulation of non-erythroid αII-spectrin and calpain-cleaved αII-spectrin breakdown products in cerebrospinal fluid after traumatic brain injury in rats. Journal of neurochemistry 78, 1297-1306 (2001). [CrossRef]
- Yokobori, S. et al. Acute diagnostic biomarkers for spinal cord injury: Review of the literature and preliminary research report. World neurosurgery 83, 867-878 (2015). [CrossRef]
- Massaro, A.N. et al. Serum biomarkers of MRI brain injury in neonatal hypoxic ischemic encephalopathy treated with whole-body hypothermia: A pilot study. Pediatric critical care medicine: A journal of the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies 14, 310 (2013). [CrossRef]
- Siman, R. et al. Evidence that the blood biomarker SNTF predicts brain imaging changes and persistent cognitive dysfunction in mild TBI patients. Frontiers in neurology 4, 190 (2013). [CrossRef]
- Ringger, N.C. et al. A novel marker for traumatic brain injury: CSF αII-spectrin breakdown product levels. Journal of neurotrauma 21, 1443-1456 (2004). [CrossRef]
- Mondello, S. et al. Increased levels of serum MAP-2 at 6-months correlate with improved outcome in survivors of severe traumatic brain injury. Brain injury 26, 1629-1635 (2012). [CrossRef]
- Papa, L. et al. Temporal profile of microtubule-associated protein 2: A novel indicator of diffuse brain injury severity and early mortality after brain trauma. Journal of neurotrauma 35, 32-40 (2018). [CrossRef]
- Korley, F.K. et al. Circulating brain-derived neurotrophic factor has diagnostic and prognostic value in traumatic brain injury. Journal of neurotrauma 33, 215-225 (2016). [CrossRef]
- Yang, J., Korley, F.K., Dai, M. & Everett, A.D. Serum neurogranin measurement as a biomarker of acute traumatic brain injury. Clinical biochemistry 48, 843-848 (2015). [CrossRef]
- Balakathiresan, N. et al. MicroRNA let-7i is a promising serum biomarker for blast-induced traumatic brain injury. Journal of neurotrauma 29, 1379-1387 (2012). [CrossRef]
- Redell, J.B., Moore, A.N., Ward III, N.H., Hergenroeder, G.W. & Dash, P.K. Human traumatic brain injury alters plasma microRNA levels. Journal of neurotrauma 27, 2147-2156 (2010). [CrossRef]
- 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. Scientific reports 6, 1-12 (2016). [CrossRef]
- Mitra, B. et al. Plasma micro-RNA biomarkers for diagnosis and prognosis after traumatic brain injury: A pilot study. Journal of Clinical Neuroscience 38, 37-42 (2017). [CrossRef]
- Agoston, D.V., Shutes-David, A. & Peskind, E.R. Biofluid biomarkers of traumatic brain injury. Brain injury 31, 1195-1203 (2017). [CrossRef]
- Moyron, R.B. et al. Differential protein expression in exosomal samples taken from trauma patients. PROTEOMICS–Clinical Applications 11, 1700061 (2017). [CrossRef]
- Manek, R. et al. Protein biomarkers and neuroproteomics characterization of microvesicles/exosomes from human cerebrospinal fluid following traumatic brain injury. Molecular neurobiology 55, 6112-6128 (2018). [CrossRef]
- Nekludov, M., Bellander, B.-M., Gryth, D., Wallen, H. & Mobarrez, F. Brain-derived microparticles in patients with severe isolated TBI. Brain injury 31, 1856-1862 (2017). [CrossRef]
- Stern, R.A. et al. Preliminary study of plasma exosomal tau as a potential biomarker for chronic traumatic encephalopathy. Journal of alzheimer's disease 51, 1099-1109 (2016). [CrossRef]
- Shi, K., Zhang, J., Dong, J.-f. & Shi, F.-D. Dissemination of brain inflammation in traumatic brain injury. Cellular & molecular immunology 16, 523-530 (2019). [CrossRef]
- Rodney, T., Osier, N. & Gill, J. Pro-and anti-inflammatory biomarkers and traumatic brain injury outcomes: A review. Cytokine 110, 248-256 (2018). [CrossRef]
- Visser, K. et al. Blood-based biomarkers of inflammation in mild traumatic brain injury: A systematic review. Neuroscience & Biobehavioral Reviews 132, 154-168 (2022). [CrossRef]
- Ooi, S.Z.Y. et al. Interleukin-6 as a prognostic biomarker of clinical outcomes after traumatic brain injury: A systematic review. Neurosurgical Review 45, 3035-3054 (2022). [CrossRef]
- Xu, L.B. et al. High-sensitivity C-reactive protein is a prognostic biomarker of six-month disability after traumatic brain injury: Results from the TRACK-TBI study. Journal of neurotrauma 38, 918-927 (2021). [CrossRef]
- Monsour, M., Ebedes, D. & Borlongan, C.V. A review of the pathology and treatment of TBI and PTSD. Experimental neurology 351, 114009 (2022). [CrossRef]
- Katrinli, S., Oliveira, N.C., Felger, J.C., Michopoulos, V. & Smith, A.K. The role of the immune system in posttraumatic stress disorder. Translational Psychiatry 12, 313 (2022). [CrossRef]
- Conroy, S.K., Brownlowe, K.B. & McAllister, T.W. Depression comorbid with stroke, traumatic brain injury, Parkinson’s disease, and multiple sclerosis: Diagnosis and treatment. Focus 18, 150-161 (2020). [CrossRef]
- Osimo, E.F., Baxter, L.J., Lewis, G., Jones, P.B. & Khandaker, G.M. Prevalence of low-grade inflammation in depression: A systematic review and meta-analysis of CRP levels. Psychological medicine 49, 1958-1970 (2019). [CrossRef]
- Pierre, K. et al. Chronic traumatic encephalopathy: Update on current clinical diagnosis and management. Biomedicines 9, 415 (2021). [CrossRef]
- Mohamed, M.S., El Sayed, I., Zaki, A. & Abdelmonem, S. Assessment of the effect of amantadine in patients with traumatic brain injury: A meta-analysis. Journal of Trauma and Acute Care Surgery 92, 605-614 (2022). [CrossRef]
- Silverberg, N.D. & Panenka, W.J. Antidepressants for depression after concussion and traumatic brain injury are still best practice. BMC psychiatry 19, 1-3 (2019). [CrossRef]
- McAllister, T.W. et al. Alpha-2 adrenergic challenge with guanfacine one month after mild traumatic brain injury: Altered working memory and BOLD response. Int J Psychophysiol 82, 107-114 (2011). [CrossRef]
- Fesharaki-Zadeh, A. Oxidative Stress in Traumatic Brain Injury. Int J Mol Sci 23 (2022). [CrossRef]
- Di Pietro, V. et al. Antioxidant Therapies in Traumatic Brain Injury. Antioxidants (Basel) 9 (2020). [CrossRef]
- Xiong, Y., Mahmood, A. & Chopp, M. Animal models of traumatic brain injury. Nature Reviews Neuroscience 14, 128-142 (2013). [CrossRef]
- Tang, S.J. et al. Fyn kinase inhibition reduces protein aggregation, increases synapse density and improves memory in transgenic and traumatic Tauopathy. Acta Neuropathologica Communications 8, 96 (2020). [CrossRef]
- Ojo, J.O. et al. Repetitive mild traumatic brain injury augments tau pathology and glial activation in aged hTau mice. J Neuropathol Exp Neurol 72, 137-151 (2013). [CrossRef]
- Weber, J.T. Experimental models of repetitive brain injuries. Prog Brain Res 161, 253-261 (2007). [CrossRef]
- Mouzon, B. et al. Repetitive mild traumatic brain injury in a mouse model produces learning and memory deficits accompanied by histological changes. J Neurotrauma 29, 2761-2773 (2012). [CrossRef]
- Huang, L. et al. Tissue vulnerability is increased following repetitive mild traumatic brain injury in the rat. Brain Res 1499, 109-120 (2013). [CrossRef]
- Petraglia, A.L. et al. The spectrum of neurobehavioral sequelae after repetitive mild traumatic brain injury: A novel mouse model of chronic traumatic encephalopathy. J Neurotrauma 31, 1211-1224 (2014). [CrossRef]
- Lucke-Wold, B.P. et al. Linking traumatic brain injury to chronic traumatic encephalopathy: Identification of potential mechanisms leading to neurofibrillary tangle development. J Neurotrauma 31, 1129-1138 (2014). [CrossRef]
- Rubenstein, R. 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 10, 124 (2019). [CrossRef]
- Lucke-Wold, B. et al. Role of Tau Acetylation in Alzheimer's Disease and Chronic Traumatic Encephalopathy: The Way Forward for Successful Treatment. J Neurol Neurosurg 4 (2017). [CrossRef]
- Min, S.W. et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med 21, 1154-1162 (2015). [CrossRef]
- Lagraoui, M. et al. Salsalate treatment following traumatic brain injury reduces inflammation and promotes a neuroprotective and neurogenic transcriptional response with concomitant functional recovery. Brain Behav Immun 61, 96-109 (2017). [CrossRef]
- Shirakawa, K. et al. Salicylate, diflunisal and their metabolites inhibit CBP/p300 and exhibit anticancer activity. Elife 5 (2016). [CrossRef]
- Tucker, D., Lu, Y. & Zhang, Q. From Mitochondrial Function to Neuroprotection-an Emerging Role for Methylene Blue. Mol Neurobiol 55, 5137-5153 (2018). [CrossRef]
- Vekaria, H.J., Talley Watts, L., Lin, A.L. & Sullivan, P.G. Targeting mitochondrial dysfunction in CNS injury using Methylene Blue; still a magic bullet? Neurochem Int 109, 117-125 (2017). [CrossRef]
- Talley Watts, L. et al. Methylene blue is neuroprotective against mild traumatic brain injury. J Neurotrauma 31, 1063-1071 (2014). [CrossRef]
- Cohen, T.J. et al. The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun 2, 252 (2011). [CrossRef]
- Rojo, A.I., Sagarra, M.R. d. & Cuadrado, A. GSK-3β down-regulates the transcription factor Nrf2 after oxidant damage: Relevance to exposure of neuronal cells to oxidative stress. Journal of neurochemistry 105, 192-202 (2008). [CrossRef]
- Rankin, C.A., Sun, Q. & Gamblin, T.C. Tau phosphorylation by GSK-3β promotes tangle-like filament morphology. Molecular neurodegeneration 2, 1-14 (2007). [CrossRef]
- Hernandez, F., Lucas, J.J. & Avila, J. GSK3 and tau: Two convergence points in Alzheimer's disease. Journal of Alzheimer's disease 33, S141-S144 (2013). [CrossRef]
- Cuadrado, A., Kügler, S. & Lastres-Becker, I. Pharmacological targeting of GSK-3 and NRF2 provides neuroprotection in a preclinical model of tauopathy. Redox biology 14, 522-534 (2018). [CrossRef]
- Dash, P.K. et al. Involvement of the glycogen synthase kinase-3 signaling pathway in TBI pathology and neurocognitive outcome. PloS one 6, e24648 (2011). [CrossRef]
- Shapira, M. et al. Role of glycogen synthase kinase-3β in early depressive behavior induced by mild traumatic brain injury. Molecular and Cellular Neuroscience 34, 571-577 (2007). [CrossRef]
- Yu, F., Zhang, Y. & Chuang, D.-M. Lithium reduces BACE1 overexpression, beta amyloid accumulation, and spatial learning deficits in mice with traumatic brain injury. Journal of neurotrauma 29, 2342-2351 (2012). [CrossRef]
- Yu, F. et al. Lithium ameliorates neurodegeneration, suppresses neuroinflammation, and improves behavioral performance in a mouse model of traumatic brain injury. Journal of neurotrauma 29, 362-374 (2012). [CrossRef]
- Yu, F. et al. Posttrauma cotreatment with lithium and valproate: Reduction of lesion volume, attenuation of blood-brain barrier disruption, and improvement in motor coordination in mice with traumatic brain injury. Journal of neurosurgery 119, 766-773 (2013). [CrossRef]
- Zhu, Z.-F., Wang, Q.-G., Han, B.-J. & William, C.P. Neuroprotective effect and cognitive outcome of chronic lithium on traumatic brain injury in mice. Brain research bulletin 83, 272-277 (2010). [CrossRef]
- Leeds, P.R. et al. A new avenue for lithium: Intervention in traumatic brain injury. ACS chemical neuroscience 5, 422-433 (2014). [CrossRef]
- Kabadi, S.V. & Faden, A.I. Selective CDK inhibitors: Promising candidates for future clinical traumatic brain injury trials. Neural Regeneration Research 9, 1578 (2014). [CrossRef]
- de Rivero Vaccari, J.P., Dietrich, W.D. & Keane, R.W. Activation and regulation of cellular inflammasomes: Gaps in our knowledge for central nervous system injury. Journal of Cerebral Blood Flow & Metabolism 34, 369-375 (2014). [CrossRef]
- Bergold, P.J. Treatment of traumatic brain injury with anti-inflammatory drugs. Experimental neurology 275, 367-380 (2016). [CrossRef]
- L. Kelso, M. & E. Gendelman, H. Bridge between neuroimmunity and traumatic brain injury. Current pharmaceutical design 20, 4284-4298 (2014).
- Adamczak, S.E. et al. Pyroptotic neuronal cell death mediated by the AIM2 inflammasome. Journal of Cerebral Blood Flow & Metabolism 34, 621-629 (2014). [CrossRef]
- Lozano, D. et al. Neuroinflammatory responses to traumatic brain injury: Etiology, clinical consequences, and therapeutic opportunities. Neuropsychiatric disease and treatment, 97-106 (2015). [CrossRef]
- Woodcock, T. & Morganti-Kossmann, M.C. The role of markers of inflammation in traumatic brain injury. Frontiers in neurology 4, 18 (2013). [CrossRef]
- Wu, H. et al. Mer regulates microglial/macrophage M1/M2 polarization and alleviates neuroinflammation following traumatic brain injury. Journal of neuroinflammation 18, 1-20 (2021). [CrossRef]
- Rodriguez-Rodriguez, A., Jose Egea-Guerrero, J., Murillo-Cabezas, F. & Carrillo-Vico, A. Oxidative stress in traumatic brain injury. Current medicinal chemistry 21, 1201-1211 (2014). [CrossRef]
- Gensel, J.C. & Zhang, B. Macrophage activation and its role in repair and pathology after spinal cord injury. Brain research 1619, 1-11 (2015). [CrossRef]
- Loane, D.J., Stoica, B.A. & Faden, A.I. Neuroprotection for traumatic brain injury. Handbook of clinical neurology 127, 343-366 (2015). [CrossRef]
- Pozdnyakov, D.I., Miroshnichenko, K.A., Voronkov, A.V. & Kovaleva, T. y. G. The administration of the new pyrimidine derivative—4-{2-[2-(3, 4-Dimethoxyphenyl)-Vinyl]-6-Ethyl-4-Oxo-5-Phenyl-4H-pyrimidine-1-Il} benzsulfamide restores the activity of brain cells in experimental chronic traumatic encephalopathy by maintaining mitochondrial function. Medicina 55, 386 (2019). [CrossRef]
- Logsdon, A.F. et al. Salubrinal reduces oxidative stress, neuroinflammation and impulsive-like behavior in a rodent model of traumatic brain injury. Brain research 1643, 140-151 (2016). [CrossRef]
- Wang, Y. et al. Calpain-2 as a therapeutic target in repeated concussion–induced neuropathy and behavioral impairment. Science Advances 6, eaba5547 (2020). [CrossRef]
- Panikashvili, D. et al. The endocannabinoid 2-AG protects the blood–brain barrier after closed head injury and inhibits mRNA expression of proinflammatory cytokines. Neurobiology of disease 22, 257-264 (2006). [CrossRef]
- Alderson, P. & Roberts, I. Corticosteroids for acute traumatic brain injury. Cochrane Database Syst Rev 2005, Cd000196 (2005). [CrossRef]
- Roberts, I. et al. Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury (MRC CRASH trial): Randomised placebo-controlled trial. Lancet 364, 1321-1328 (2004). [CrossRef]
- Hein, A.M. & O’Banion, M.K. Neuroinflammation and memory: The role of prostaglandins. Molecular neurobiology 40, 15-32 (2009). [CrossRef]
- Shohami, E., Gallily, R., Mechoulam, R., Bass, R. & Ben-Hur, T. Cytokine production in the brain following closed head injury: Dexanabinol (HU-211) is a novel TNF-α inhibitor and an effective neuroprotectant. Journal of neuroimmunology 72, 169-177 (1997). [CrossRef]
- Chio, C.-C. et al. Etanercept attenuates traumatic brain injury in rats by reducing early microglial expression of tumor necrosis factor-α. BMC neuroscience 14, 1-12 (2013). [CrossRef]
- Finnie, J. Neuroinflammation: Beneficial and detrimental effects after traumatic brain injury. Inflammopharmacology 21, 309-320 (2013). [CrossRef]
- Tehranian, R. et al. Improved recovery and delayed cytokine induction after closed head injury in mice with central overexpression of the secreted isoform of the interleukin-1 receptor antagonist. Journal of neurotrauma 19, 939-951 (2002). [CrossRef]
- Anderson, G.D. et al. Comparison of the effects of erythropoietin and anakinra on functional recovery and gene expression in a traumatic brain injury model. Frontiers in Pharmacology 4, 129 (2013). [CrossRef]
- Helmy, A. et al. Recombinant human interleukin-1 receptor antagonist in severe traumatic brain injury: A phase II randomized control trial. Journal of Cerebral Blood Flow & Metabolism 34, 845-851 (2014). [CrossRef]
- Atkins, C.M., Cepero, M.L., Kang, Y., Liebl, D.J. & Dietrich, W.D. Effects of early rolipram treatment on histopathological outcome after controlled cortical impact injury in mice. Neuroscience letters 532, 1-6 (2013). [CrossRef]
- Atkins, C. et al. Postinjury treatment with rolipram increases hemorrhage after traumatic brain injury. Journal of neuroscience research 90, 1861-1871 (2012). [CrossRef]
- Garrido-Mesa, N., Zarzuelo, A. & Galvez, J. What is behind the non-antibiotic properties of minocycline? Pharmacological research 67, 18-30 (2013). [CrossRef]
- Abdel Baki, S.G., Schwab, B., Haber, M., Fenton, A.A. & Bergold, P.J. Minocycline synergizes with N-acetylcysteine and improves cognition and memory following traumatic brain injury in rats. PloS one 5, e12490 (2010). [CrossRef]
- Adembri, C. et al. Minocycline but not tigecycline is neuroprotective and reduces the neuroinflammatory response induced by the superimposition of sepsis upon traumatic brain injury. Critical care medicine 42, e570-e582 (2014). [CrossRef]
- Haber, M. et al. Minocycline plus N-acetylcysteine synergize to modulate inflammation and prevent cognitive and memory deficits in a rat model of mild traumatic brain injury. Experimental neurology 249, 169-177 (2013). [CrossRef]
- Homsi, S. et al. Blockade of acute microglial activation by minocycline promotes neuroprotection and reduces locomotor hyperactivity after closed head injury in mice: A twelve-week follow-up study. Journal of neurotrauma 27, 911-921 (2010). [CrossRef]
- Wei, J. & Xiao, G.-m. The neuroprotective effects of progesterone on traumatic brain injury: Current status and future prospects. Acta Pharmacologica Sinica 34, 1485-1490 (2013). [CrossRef]
- Sarkaki, A.R., Khaksari Haddad, M., Soltani, Z., Shahrokhi, N. & Mahmoodi, M. Time-and dose-dependent neuroprotective effects of sex steroid hormones on inflammatory cytokines after a traumatic brain injury. Journal of neurotrauma 30, 47-54 (2013). [CrossRef]
- Wright, D.W. et al. ProTECT: A randomized clinical trial of progesterone for acute traumatic brain injury. Annals of emergency medicine 49, 391-402. e392 (2007). [CrossRef]
- Wright, D.W. et al. Very early administration of progesterone for acute traumatic brain injury. New England Journal of Medicine 371, 2457-2466 (2014). [CrossRef]
- Skolnick, B.E. et al. A clinical trial of progesterone for severe traumatic brain injury. New England Journal of Medicine 371, 2467-2476 (2014). [CrossRef]
- Radosevich, J.J., Patanwala, A.E. & Erstad, B.L. Emerging pharmacological agents to improve survival from traumatic brain injury. Brain injury 27, 1492-1499 (2013). [CrossRef]
- Bian, X.-x., Yuan, X.-s. & Qi, C.-p. Effect of recombinant human erythropoietin on serum S100B protein and interleukin-6 levels after traumatic brain injury in the rat. Neurologia medico-chirurgica 50, 361-366 (2010). [CrossRef]
- Hellewell, S.C., Yan, E.B., Alwis, D.S., Bye, N. & Morganti-Kossmann, M.C. Erythropoietin improves motor and cognitive deficit, axonal pathology, and neuroinflammation in a combined model of diffuse traumatic brain injury and hypoxia, in association with upregulation of the erythropoietin receptor. Journal of neuroinflammation 10, 1-21 (2013). [CrossRef]
- Robertson, C.S. et al. Effect of erythropoietin and transfusion threshold on neurological recovery after traumatic brain injury: A randomized clinical trial. Jama 312, 36-47 (2014). [CrossRef]
- Talving, P. et al. Erythropoiesis-stimulating agent administration and survival after severe traumatic brain injury: A prospective study. Arch Surg 147, 251-255 (2012). [CrossRef]
- Schneider, E.B. et al. Premorbid statin use is associated with improved survival and functional outcomes in older head-injured individuals. J Trauma 71, 815-819 (2011). [CrossRef]
- Sacramento, C.B. et al. Anti-phospho-tau gene therapy for chronic traumatic encephalopathy. Human gene therapy 31, 57-69 (2020). [CrossRef]
- Nobuhara, C.K. et al. Tau antibody targeting pathological species blocks neuronal uptake and interneuron propagation of tau in vitro. The American journal of pathology 187, 1399-1412 (2017). [CrossRef]
- Kondo, A. et al. Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature 523, 431-436 (2015). [CrossRef]
- Lu, K.P. et al. Potential of the antibody against cis–phosphorylated tau in the early diagnosis, treatment, and prevention of Alzheimer disease and brain injury. JAMA neurology 73, 1356-1362 (2016). [CrossRef]
- Houck, A., Kondo, A. & Lu, K.P. (AAN Enterprises, 2017).
- Willis, E.F. et al. Repopulating microglia promote brain repair in an IL-6-dependent manner. Cell 180, 833-846. e816 (2020). [CrossRef]
- Shaito, A. et al. Western diet aggravates neuronal insult in post-traumatic brain injury: Proposed pathways for interplay. EBioMedicine 57, 102829 (2020). [CrossRef]
- Nguyen, J.C., Killcross, A.S. & Jenkins, T.A. Obesity and cognitive decline: Role of inflammation and vascular changes. Frontiers in neuroscience 8, 375 (2014). [CrossRef]
- Posey, K.A. et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. American Journal of Physiology-Endocrinology and Metabolism 296, E1003-E1012 (2009). [CrossRef]
- Gemma, C. & Bickford, P.C. Interleukin-1ß and Caspase-1: Players in the regulation of age-related cognitive dysfunction. Reviews in the neurosciences 18, 137-148 (2007). [CrossRef]
- Sherman, M. et al. Adult obese mice suffer from chronic secondary brain injury after mild TBI. Journal of neuroinflammation 13, 1-10 (2016). [CrossRef]
- Avena, N.M. & Gold, M.S. Food and addiction–sugars, fats and hedonic overeating. Addiction 106, 1214-1215 (2011). [CrossRef]
- Zilberter, T. & Zilberter, Y. Ketogenic ratio determines metabolic effects of macronutrients and prevents interpretive bias. Frontiers in nutrition 5, 75 (2018). [CrossRef]
- Gasior, M., Rogawski, M.A. & Hartman, A.L. Neuroprotective and disease-modifying effects of the ketogenic diet. Behavioural pharmacology 17, 431 (2006). [CrossRef]
- Murphy, P., Likhodii, S., Nylen, K. & Burnham, W. The antidepressant properties of the ketogenic diet. Biological psychiatry 56, 981-983 (2004). [CrossRef]
- Krikorian, R. et al. Dietary ketosis enhances memory in mild cognitive impairment. Neurobiology of aging 33, 425. e419-425. e427 (2012). [CrossRef]
- White, H. & Venkatesh, B. Clinical review: Ketones and brain injury. Critical care 15, 1-10 (2011). [CrossRef]
- Anastasiou, C.A. et al. Mediterranean diet and cognitive health: Initial results from the Hellenic Longitudinal Investigation of Ageing and Diet. PloS one 12, e0182048 (2017). [CrossRef]
- McGrattan, A.M. et al. Diet and inflammation in cognitive ageing and Alzheimer’s disease. Current nutrition reports 8, 53-65 (2019). [CrossRef]
- Ritter, A., Shan, G., Montes, A., Randall, R. & Bernick, C. Traumatic encephalopathy syndrome: Application of new criteria to a cohort exposed to repetitive head impacts. British journal of sports medicine 57, 389-394 (2023). [CrossRef]
- Ruprecht, R., Scheurer, E. & Lenz, C. Systematic review on the characterization of chronic traumatic encephalopathy by MRI and MRS. Journal of Magnetic Resonance Imaging 49, 212-228 (2019). [CrossRef]
- Goldstein, L.E., McKee, A.C. & Stanton, P.K. Considerations for animal models of blast-related traumatic brain injury and chronic traumatic encephalopathy. Alzheimer's research & therapy 6, 1-10 (2014). [CrossRef]
- Fitzgerald, N.D. & Cammack, K.M. Chronic Traumatic Encephalopathy: A Review of Clinical Diagnosis, Animal Models, Sex Differences, and A Revised Return-to-Play Protocol. (2017).
- Risling, M. et al. Modelling human pathology of traumatic brain injury in animal models. Journal of internal medicine 285, 594-607 (2019). [CrossRef]
- Fesharaki-Zadeh, A., Miyauchi, J.T., St. Laurent-Arriot, K., Tsirka, S.E. & Bergold, P.J. Increased Behavioral Deficits and Inflammation in a Mouse Model of Co-Morbid Traumatic Brain Injury and Post-Traumatic Stress Disorder. ASN neuro 12, 1759091420979567 (2020). [CrossRef]
- Breen, P.W. & Krishnan, V. Recent preclinical insights into the treatment of chronic traumatic encephalopathy. Frontiers in Neuroscience 14, 616 (2020). [CrossRef]
- Lindsley, C.W. Vol. 8 1629-1631 (ACS Publications, 2017).
- Fesharaki-Zadeh, A. Chronic traumatic encephalopathy: A brief overview. Frontiers in neurology, 713 (2019). [CrossRef]
- Nylen, K. et al. Increased serum-GFAP in patients with severe traumatic brain injury is related to outcome. Journal of the neurological sciences 240, 85-91 (2006). [CrossRef]
- Shahim, P. et al. Serum neurofilament light protein predicts clinical outcome in traumatic brain injury. Scientific reports 6, 36791 (2016). [CrossRef]
- Abbasi, M., Sajjadi, M., Fathi, M. & Maghsoudi, M. Serum S100B protein as an outcome prediction tool in emergency department patients with traumatic brain injury. Turkish journal of emergency medicine 14, 147-152 (2014). [CrossRef]
- Bazarian, J.J., Zemlan, F.P., Mookerjee, S. & Stigbrand, T. Serum S-100B and cleaved-tau are poor predictors of long-term outcome after mild traumatic brain injury. Brain injury 20, 759-765 (2006). [CrossRef]
- McKee, A.C. et al. Chronic traumatic encephalopathy (CTE): Criteria for neuropathological diagnosis and relationship to repetitive head impacts. Acta neuropathologica, 1-24 (2023). [CrossRef]



| Serum Biomarker | TBI Outcomes |
|---|---|
| GFAP | Clinical TBI studies have reported longitudinal elevation in GFAP levels 27. GFAP was also recently approved by the FDA as a TBI outcome clinical measure 47. |
| NfL | Clinical TBI studies have reported elevated NfL serum levels both acutely and longitudinally27. |
| Tau (total) | Total tau elevation has been reported both acutely and chronically in TBI populations 27. |
| NSE | NSE elevated levels have reported in both mild and more severe TBI populations31,32. |
| UCHL-1 | UCHL-1 has been shown to be robustly elevated in both mTBI and more severe TBI patients 134. UCHL-1 was recently FDA approved as a TBI outcome clinical measure 47. |
| S100B | S100B has been reported to be more acutely elevated in various TBI severity cases 48,49. |
| SBDP | SBDPs are products of calpain and caspase-3 post TBI, and have been reported to be elevated in both preclinical and clinical studies 56,65. |
| MBP | MBP is an oligodentrocyte protein and a product of proteases including calpain and reported to be elevated in severe TBI patients 43,64. |
| MAP-2 | An emerging biomarker for TBI patients39. |
| BDNF | Mainly reported in the preclinical TBI studies, with potential application to clinical TBI population67. |
| microRNA | a class of small endogenous RNA molecules, and has been reported to be elevated in biofluid (CSF, serum, or plasma) in several rodent models of TBI of various severities69. |
| MV/E | lipid-bilayered, encapsulated particles (10–100 nm in diameter) that are released from cells into the CSF and blood during TBI73. reported elevated MV/E released into CSF in TBI patients74. |
|
Proinflammatory cytokines (IL-6, IL-1, IL-8, IL-10, TNFα, CRP) |
Proinflammatory markers, especially Il-6 and CRP have been shown to have robust diagnostic and prognostic value (ref). |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).