An update on diagnostic and prognostic biomarkers for traumatic brain injury

Kevin K Wang, Zhihui Yang, Tian Zhu, Yuan Shi, Richard Rubenstein, J Adrian Tyndall, Geoff T Manley, Kevin K Wang, Zhihui Yang, Tian Zhu, Yuan Shi, Richard Rubenstein, J Adrian Tyndall, Geoff T Manley

Abstract

Traumatic brain injury (TBI) is a major worldwide neurological disorder of epidemic proportions. To date, there are still no FDA-approved therapies to treat any forms of TBI. Encouragingly, there are emerging data showing that biofluid-based TBI biomarker tests have the potential to diagnose the presence of TBI of different severities including concussion, and to predict outcome. Areas covered: The authors provide an update on the current knowledge of TBI biomarkers, including protein biomarkers for neuronal cell body injury (UCH-L1, NSE), astroglial injury (GFAP, S100B), neuronal cell death (αII-spectrin breakdown products), axonal injury (NF proteins), white matter injury (MBP), post-injury neurodegeneration (total Tau and phospho-Tau), post-injury autoimmune response (brain antigen-targeting autoantibodies), and other emerging non-protein biomarkers. The authors discuss biomarker evidence in TBI diagnosis, outcome prognosis and possible identification of post-TBI neurodegernative diseases (e.g. chronic traumatic encephalopathy and Alzheimer's disease), and as theranostic tools in pre-clinical and clinical settings. Expert commentary: A spectrum of biomarkers is now at or near the stage of formal clinical validation of their diagnostic and prognostic utilities in the management of TBI of varied severities including concussions. TBI biomarkers could serve as a theranostic tool in facilitating drug development and treatment monitoring.

Keywords: Biomarkers; diagnostics; neurotrauma; theranostics; traumatic brain injury.

Figures

Figure 1.. Graphic representation of major TBI…
Figure 1.. Graphic representation of major TBI protein biomarkers linked to different pathophysiologic processes in TBI.
These processes thus far include axonal injury, dendritic injury, neuronal cell body injury, demyelination, synaptic injury and astroglia injury and microglia responses. Cellular and subcellular localization of representative TBI biomarkers are also shown with immunocytochemical staining images (based on mouse brain data).
Figure 2.. A continuum of protein biomarkers…
Figure 2.. A continuum of protein biomarkers in tracking different phases of TBI.
Acute neuronal cell body injury markers UCH-L1 (ubiquitin C-terminal hydrolase-L1), NSE (neuronal specific enolase), necrosis markers SBDP150/145 (αII-spectrin breakdown product 150 kDa & 145 kDa), SNTF (αII-spectrin N-terminal fragment), S100B (glial calcium-binding protein S100B), GFAP & BDP (glial fibrillary acidic protein & breakdown product), delayed axonal injury NF-H, M, L (neurofilament-heavy, medium & light), demyelination marker MBP (myelin basic protein), apoptosis marker SBDP120 (αII-spectrin breakdown product 120 kDa), autoimmunity markers auto[GFAP] (autoantibodies to GFAP), neurodegeneration markers Tau (tau protein), P-Tau (phosphorylated tau), Aβ (amyloid β-peptides) and TDP-43.

References

    1. Hoffman SW, Harrison C. The interaction between psychological health and traumatic brain injury: a neuroscience perspective. The Clinical Neuropsychologist. 23(8), 1400–1415 (2009).
    1. Centers for Disease, Control, Prevention. CDC grand rounds: reducing severe traumatic brain injury in the United States. MMWR Morb. Mortal. Wkly. Rep. 62(27), 549–552 (2013).
    1. DOD-DVBIC. DVBIC Worldwide-TBI 2000–2013 Report. DOD Report. 1–5 (2014).
    1. Elovic E, Doppalapudi HS, Miller M. Endocrine abnormalities and fatigue after traumatic brain injury. J Head Trauma Rehab. 21(5), 426–427 (2006).
    1. Englander J, Bushnik T, Oggins J, Katznelson L. Fatigue after traumatic brain injury: Association with neuroendocrine, sleep, depression and other factors. Brain Inj. 24(12), 1379–1388 (2010).
    1. Maas AI, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. The Lancet Neurology. 7(8), 728–741 (2008).
    1. Kibby MY, Long CJ. Minor head injury: attempts at clarifying the confusion. Brain Inj. 10(3), 159–186 (1996).
    1. Pearson WS, Sugerman DE, McGuire LC, Coronado VG. Emergency department visits for traumatic brain injury in older adults in the United States: 2006–08. West J Emerg Med. 13(3), 289–293 (2012).
    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, e55–71 (2013).
    1. Langlois JA, Rutland-Brown W, Thomas KE. Traumatic Brain Injury in the United States: Emergency Department Visits, Hospitalizations, and Deaths. PsycEXTRA Dataset.
    1. Pellman EJ, Viano DC, Casson IR, et al. Concussion in professional football: repeat injuries--part 4. Neurosurgery. 55(4), 860–73– discussion 873–6 (2004).
    1. Slobounov S, Slobounov E, Sebastianelli W, Cao C, Newell K. Differential rate of recovery in athletes after first and second concussion episodes. Neurosurgery. 61(2), 338–44– discussion 344 (2007).
    1. Dambinova SA, Bettermann K, Glynn T, et al. Diagnostic potential of the NMDA receptor peptide assay for acute ischemic stroke. PLoS ONE. 7(7), e42362 (2012).
    1. Papa L, Akinyi L, Liu MC, et al. Ubiquitin C-terminal hydrolase is a novel biomarker in humans for severe traumatic brain injury. Crit. Care Med. 38(1), 138–144 (2010).
    1. Saatman KE, Duhaime A-C, Bullock R, Maas AIR, Valadka A, Manley GT. Classification of Traumatic Brain Injury for Targeted Therapies. J. Neurotrauma. 25(7), 719–738 (2008).
    1. Smith DH, Meaney DF, Shull WH. Diffuse axonal injury in head trauma. The Journal of Head Trauma Rehabilitation. 18(4), 307–316 (2003).
    1. Xu JJ, Rasmussen I-AI, Lagopoulos JJ, Håberg AA. Diffuse axonal injury in severe traumatic brain injury visualized using high-resolution diffusion tensor imaging. J. Neurotrauma. 24(5), 753–765 (2007).
    1. Merlo L, Cimino F, Angileri FF, et al. Alteration in synaptic junction proteins following traumatic brain injury. J. Neurotrauma. 31(16), 1375–1385 (2014).
    1. Winston CN, Noël A, Neustadtl A, et al. Dendritic Spine Loss and Chronic White Matter Inflammation in a Mouse Model of Highly Repetitive Head Trauma. Am. J. Pathol. 186(3), 552–567 (2016).
    1. Zhang Z, Moghieb A, Kevin K Wang W. Acute, Subacute and chronic Biomarkers for CNS injury In: Biomarkers of Brain Injury and Neurological Disorders. Wang KKW, Zhang Z, Kobeissy FH (Eds.). CRC Press, 1–25 (2014).
    1. Wang KK, Moghieb A, Yang Z, Zhang Z. Systems biomarkers as acute diagnostics and chronic monitoring tools for traumatic brain injury. SPIE Defense, Security, and Sensing. 87230O–87230O–15 (2013).
    1. Mondello S, Muller U, Jeromin A, Streeter J, Hayes RL, Wang KKW. Blood-based diagnostics of traumatic brain injuries. Expert Rev. Mol. Diagn. 11(1), 65–78 (2011).
    1. Zetterberg H, Blennow K. Fluid biomarkers for mild traumatic brain injury and related conditions. Nature Reviews Neurology. 12(10), 563–574 (2016).
    1. Bohmer AE, Oses JP, Schmidt AP, et al. Neuron-specific enolase, S100B, and glial fibrillary acidic protein levels as outcome predictors in patients with severe traumatic brain injury. Neurosurgery. 68(6), 1624–30– discussion 1630–1 (2011).
    1. Stein DM, Kufera JA, Lindell A, et al. Association of CSF Biomarkers and Secondary Insults Following Severe Traumatic Brain Injury. Neurocrit Care. 14(2), 200–207 (2011).
    1. Berger RP, Pierce MC, Wisniewski SR, et al. Neuron-specific enolase and S100B in cerebrospinal fluid after severe traumatic brain injury in infants and children. PEDIATRICS. 109(2), E31 (2002).
    1. Liu M, Zhang C, Liu W, et al. A novel rat model of blast-induced traumatic brain injury simulating different damage degree: implications for morphological, neurological, and biomarker changes. Front Cell Neurosci. 9, 168 (2015).
    1. Topolovec-Vranic J, Pollmann-Mudryj M-A, Ouchterlony D, et al. The value of serum biomarkers in prediction models of outcome after mild traumatic brain injury. J Trauma [Internet]. 71(5 Suppl 1), S478–86 (2011).
    1. Buonora JE, Yarnell AM, Lazarus RC, et al. Multivariate Analysis of Traumatic Brain Injury: Development of an Assessment Score. Front Neurol. 6(Suppl 1), 503 (2015).
    1. Agoston DV, Elsayed M. Serum-based protein biomarkers in blast-induced traumatic brain injury spectrum disorder. Front Neurol. 3, 107 (2012).
    1. Verfaillie CJ, Delanghe JR. Hemolysis correction factor in the measurement of serum neuron-specific enolase. Clin. Chem. Lab. Med. 48(6) (2010).
    1. Kobeissy FH, Ottens AK, Zhang Z, et al. Novel differential neuroproteomics analysis of traumatic brain injury in rats. Molecular \& Cellular Proteomics [Internet]. 5(10), 1887–1898 (2006).

      * Original paper describing the identification of UCH-L1 as a potential TBI biomarker

    1. Liu MC, Akinyi L, Scharf D, et al. Ubiquitin C-terminal hydrolase-L1 as a biomarker for ischemic and traumatic brain injury in rats. Eur. J. Neurosci. 31(4), 722–732 (2010).

      * First paper using ELISA assay to show UCH-L1 elevations in biofluid in rat model of TBI.

    1. Mondello S, Linnet A, Büki A, et al. Clinical utility of serum levels of ubiquitin C-terminal hydrolase as a biomarker for severe traumatic brain injury. Neurosurgery [Internet]. 70(3), 666–675 (2012).

      * Key paper describing UCH-L1 as a potential severe TBI biomarker

    1. Brophy GM, Mondello S, Papa L, et al. Biokinetic analysis of ubiquitin C-terminal hydrolase-L1 (UCH-L1) in severe traumatic brain injury patient biofluids. J. Neurotrauma. 28(6), 861–870 (2011).
    1. Papa L, Oli MW, Akinyi L, et al. UCH-L1 is a novel biomarker for severe traumatic brain injury in human. Critical Care Med. 38(1), 138–144 (2010).
    1. Zhang Z-Y, Zhang L-X, Dong X-Q, 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).
    1. Diaz-Arrastia R, Wang KKW, Papa L, et al. Acute Biomarkers of Traumatic Brain Injury: Relationship between Plasma Levels of Ubiquitin C-Terminal Hydrolase-L1 and Glial Fibrillary Acidic Protein | Abstract. J. Neurotrauma. 31(1), 19–25 (2014).
    1. Welch RD, Ayaz SI, Lewis LM, et al. Ability of Serum Glial Fibrillary Acidic Protein, Ubiquitin C-Terminal Hydrolase-L1, and S100B To Differentiate Normal and Abnormal Head Computed Tomography Findings in Patients with Suspected Mild or Moderate Traumatic Brain Injury. J. Neurotrauma. 33(2), 203–214 (2016).
    1. Tate CM, Wang KKW, Eonta S, et al. Serum brain biomarker level, neurocognitive performance, and self-reported symptom changes in soldiers repeatedly exposed to low-level blast: a breacher pilot study. J. Neurotrauma. 30(19), 1620–1630 (2013).
    1. Puvenna V, Brennan C, Shaw G, et al. Significance of Ubiquitin Carboxy-Terminal Hydrolase L1 Elevations in Athletes after Sub-Concussive Head Hits. PLoS ONE. 9(5), e96296 (2014).

      * UCH-L1 as a possible biomarker for sports-related concussions.

    1. Schulte S, Podlog LW, Hamson-Utley JJ, Strathmann FG, Strüder HK. A systematic review of the biomarker S100B: implications for sport-related concussion management. J Athl Train. 49(6), 830–850 (2014).
    1. Filippidis AS, Papadopoulos DC, Kapsalaki EZ, Fountas KN. Role of the S100B serum biomarker in the treatment of children suffering from mild traumatic brain injury. Neurosurg Focus. 29(5), E2 (2010).
    1. Ringger NC, O’steen BE, Brabham JG, et al. A novel marker for traumatic brain injury: CSF alphaII-spectrin breakdown product levels. J. Neurotrauma. 21(10), 1443–1456 (2004).
    1. Kiechle K, Bazarian JJ, Merchant-Borna K, et al. Subject-Specific Increases in Serum S-100B Distinguish Sports-Related Concussion from Sports-Related Exertion. PLoS ONE. 9(1), e84977 (2014).
    1. Vos PE, Jacobs B, Andriessen TM, et al. GFAP and S100B are biomarkers of traumatic brain injury: an observational cohort study. Neurology [Internet]. 75(20), 1786–1793 (2010).
    1. Cervellin G, Benatti M, Carbucicchio A, et al. Serum levels of protein S100B predict intracranial lesions in mild head injury. Clin. Biochem. 45(6), 408–411 (2012).
    1. Papa L, Silvestri S, Brophy GM, 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. J. Neurotrauma. 31(22), 1815–1822 (2014).
    1. Metting Z, Wilczak N, Rodiger LA, Schaaf JM, van der Naalt J. GFAP and S100B in the acute phase of mild traumatic brain injury. Neurology. 78(18), 1428–1433 (2012).
    1. Barbosa RR, Jawa R, Watters JM, et al. Evaluation and management of mild traumatic brain injury. Journal of Trauma and Acute Care Surgery. 73, S307–S314 (2012).
    1. Zongo D, Ribéreau-Gayon R, Masson F, et al. S100-B protein as a screening tool for the early assessment of minor head injury. Ann Emerg Med. 59(3), 209–218 (2012).
    1. Zhang Z, Zoltewicz JS, Mondello S, et al. Human traumatic brain injury induces autoantibody response against glial fibrillary acidic protein and its breakdown products. PLoS ONE. 9(3), e92698 (2014).

      ** First paper describing anti-GFAP autoantibody as a dominant post-TBI autoimmune response.

    1. Nylén K, Öst M, Csajbok LZ, et al. Increased serum-GFAP in patients with severe traumatic brain injury is related to outcome. J. Neurol. Sci. 240(1–2), 85–91 (2006).
    1. Mondello S, Jeromin A, Buki A, et al. Glial neuronal ratio: a novel index for differentiating injury type in patients with severe traumatic brain injury. J. Neurotrauma. 29(6), 1096–1104 (2012).
    1. Vos PE, Lamers KJ, Hendriks JC, et al. Glial and neuronal proteins in serum predict outcome after severe traumatic brain injury. Neurology. 62(8), 1303–1310 (2004).
    1. Okonkwo DO, Yue JK, Puccio AM, et al. GFAP-BDP as an acute diagnostic marker in traumatic brain injury: results from the prospective transforming research and clinical knowledge in traumatic brain injury study. J. Neurotrauma. 30(17), 1490–1497 (2013).

      ** First paper describing GFAP / GFAP-BDP as diagnostic and prognostic biomarker for full spectrum of TBI severity.

    1. Yang Z, Wang KKW. Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker. Trends in neurosciences. 38(6), 364–374 (2015).
    1. Glushakova OY, Jeromin A, Martinez J, et al. Cerebrospinal fluid protein biomarker panel for assessment of neurotoxicity induced by kainic acid in rats. Toxicological Sciences. 130(1), 158–167 (2012).
    1. Zoltewicz JS, Mondello S, Yang B, et al. Biomarkers track damage after graded injury severity in a rat model of penetrating brain injury. J. Neurotrauma. 30(13), 1161–1169 (2013).
    1. Svetlov SI, Prima V, Kirk DR, et al. Neuro-Glial and Systemic Mechanisms of Pathological Responses to Primary Blast Overpressure (OP) Compared to Severe “Composite”Blast in Rat Models. NATO Research and Technology Organisation; RTO-MP-HFM. 207, 37–1 – 37–10 (2012).
    1. Ahmed F, Gyorgy A, Kamnaksh A, et al. Time-dependent changes of protein biomarker levels in the cerebrospinal fluid after blast traumatic brain injury. Electrophoresis. 33(24), 3705–3711 (2012).
    1. Liu M-D, Luo P, Wang Z-J, Fei Z. Changes of serum Tau, GFAP, TNF-α and malonaldehyde after blast-related traumatic brain injury. Chin J Traumatol. 17(6), 317–322 (2014).
    1. Kou Z, Gattu R, Kobeissy F, et al. Combining biochemical and imaging markers to improve diagnosis and characterization of mild traumatic brain injury in the acute setting: results from a pilot study. PLoS ONE. 8(11), e80296 (2013).
    1. Schiff LL, Hadker NN, Weiser SS, Rausch CC. A literature review of the feasibility of glial fibrillary acidic protein as a biomarker for stroke and traumatic brain injury. Mol Diagn Ther. 16(2), 79–92 (2012).
    1. Pike BR, Flint J, Dutta S, Johnson E, Wang KK, Hayes RL. Accumulation of non-erythroid alpha II-spectrin and calpain-cleaved alpha II-spectrin breakdown products in cerebrospinal fluid after traumatic brain injury in rats. J. Neurochem. 78(6), 1297–1306 (2001).
    1. Mondello S, Robicsek SA, Gabrielli A, et al. alphaII-spectrin breakdown products (SBDPs): diagnosis and outcome in severe traumatic brain injury patients. J. Neurotrauma. 27(7), 1203–1213 (2010).
    1. Vartanian MG, Cordon JJ, Kupina NC, et al. Phenytoin pretreatment prevents hypoxic-ischemic brain damage in neonatal rats. Developmental brain research [Internet]. 95(2), 169–175 (1996).
    1. Berger RP, Houle J-F, Hayes RL, Wang KK, Mondello S, Bell MJ. Translating biomarkers research to clinical care: applications and issues for rehabilomics. PM & R. 3(6 Suppl 1), S31–8 (2011).
    1. Pike BR, Flint J, Dave JR, et al. Accumulation of calpain and caspase-3 proteolytic fragments of brain-derived alphaII-spectrin in cerebral spinal fluid after middle cerebral artery occlusion in rats. J. Cereb. Blood Flow Metab. 24(1), 98–106 (2004).
    1. Yokobori S, Zhang Z, Moghieb A, et al. Acute Diagnostic Biomarkers for Spinal Cord Injury: Review of the Literature and Preliminary Research Report. World Neurosurgery. 83(5), 867–878 (2015).
    1. Massaro AN, Jeromin A, Kadom N, et al. Serum Biomarkers of MRI Brain Injury in Neonatal Hypoxic Ischemic Encephalopathy Treated With Whole-Body Hypothermia. Pediatr Crit Care Med. 14(3), 310–317 (2013).
    1. Papa L, Robertson CS, Wang KKW, et al. Biomarkers improve clinical outcome predictors of mortality following non-penetrating severe traumatic brain injury. Neurocrit Care. 22(1), 52–64 (2014).
    1. Siman R Evidence that the blood biomarker SNTF predicts brain imaging changes and persistent cognitive dysfunction in mild TBI patients. 1–8 (2013).
    1. Posmantur RM, Zhao X, Kampfl A, Clifton GL, Hayes RL. Immunoblot analyses of the relative contributions of cysteine and aspartic proteases to neurofilament breakdown products following experimental brain injury in rats. Neurochemical research [Internet]. 23(10), 1265–1276 (1998).
    1. Posmantur R, Hayes RL, Dixon CE, Taft WC. Neurofilament 68 and neurofilament 200 protein levels decrease after traumatic brain injury. J. Neurotrauma. 11(5), 533–545 (1994).
    1. Anderson KJ, Scheff SW, Miller KM, et al. The phosphorylated axonal form of the neurofilament subunit NF-H (pNF-H) as a blood biomarker of traumatic brain injury. J. Neurotrauma [Internet]. 25(9), 1079–1085 (2008).
    1. Li Y, Zhang L, Kallakuri S, Cohen A, Cavanaugh JM. Correlation of mechanical impact responses and biomarker levels: A new model for biomarker evaluation in TBI. J. Neurol. Sci. 359(1–2), 280–286 (2015).
    1. Martínez-Morillo E, Childs C, García BP, et al. Neurofilament medium polypeptide (NFM) protein concentration is increased in CSF and serum samples from patients with brain injury. Clinical Chemical Laboratory Medicine. 53(10), 1575–1584 (2015).
    1. Neselius S, Zetterberg H, Blennow K, Marcusson J, Brisby H. Increased CSF levels of phosphorylated neurofilament heavy protein following bout in amateur boxers. PLoS ONE. 8(11), e81249 (2013).
    1. Žurek J, Bartlová L, Fedora M. Hyperphosphorylated neurofilament NF-H as a predictor of mortality after brain injury in children. Brain Inj. 25(2), 221–226 (2011).
    1. Oliver JM, Jones MT, Kirk KM, et al. Serum Neurofilament Light in American Football Athletes over the Course of a Season. J. Neurotrauma. 33(19), 1784–1789 (2016).
    1. Nimer Al F, Thelin E, Nyström H, et al. Comparative Assessment of the Prognostic Value of Biomarkers in Traumatic Brain Injury Reveals an Independent Role for Serum Levels of Neurofilament Light. PLoS ONE. 10(7), e0132177 (2015).
    1. Deber CM, Reynolds SJ. Central nervous system myelin: structure, function, and pathology. Clin. Biochem. 24(2), 113–134 (1991).
    1. Ottens AK, Golden EC, Bustamante L, Hayes RL, Denslow ND, Wang KKW. Proteolysis of multiple myelin basic protein isoforms after neurotrauma: characterization by mass spectrometry. J. Neurochem. 104(5), 1404–1414 (2008).
    1. Liu MC, Akle V, Zheng W, et al. Extensive degradation of myelin basic protein isoforms by calpain following traumatic brain injury. J. Neurochem. 98(3), 700–712 (2006).
    1. Wilkinson CW, Pagulayan KF, Petrie EC, et al. High prevalence of chronic pituitary and target-organ hormone abnormalities after blast-related mild traumatic brain injury. Front Neurol. 3, 11 (2012).
    1. Ponsford JL, Ziino C, Parcell DL, et al. Fatigue and sleep disturbance following traumatic brain injury--their nature, causes, and potential treatments. The Journal of Head Trauma Rehabilitation. 27(3), 224–233 (2012).
    1. Wagner AK, Zitelli KT. A Rehabilomics focused perspective on molecular mechanisms underlying neurological injury, complications, and recovery after severe TBI. Pathophysiology. 20(1), 39–48 (2013).
    1. Jordan BD. The clinical spectrum of sport-related traumatic brain injury. Nat Clin Pract Neurol. – (2013).
    1. Gavett BE, Stern RA, McKee AC. Chronic traumatic encephalopathy: a potential late effect of sport-related concussive and subconcussive head trauma. Clin Sports Med. 30(1), 179–88– xi (2011).
    1. Sorokina EGE, Semenova ZBZ, Granstrem OKO, et al. [S100B protein and autoantibodies to S100B protein in diagnostics of brain damage in craniocerebral trauma in children]. Zh Nevrol Psikhiatr Im S S Korsakova. 110(8), 30–35 (2010).
    1. Mecocci P, Parnetti L, Romano G, et al. Serum anti-GFAP and anti-S100 autoantibodies in brain aging, Alzheimer’s disease and vascular dementia. J Neuroimmunol [Internet]. 57(1–2), 165–170 (1995).
    1. Gordon WA, Zafonte R, Cicerone K, et al. Traumatic brain injury rehabilitation: state of the science. Am J Phys Med Rehabil. 85(4), 343–382 (2006).
    1. Ziebell JM, Morganti-Kossmann MC. Involvement of Pro- and Anti-Inflammatory Cytokines and Chemokines in the Pathophysiology of Traumatic Brain Injury. Neurotherapeutics [Internet]. 7(1), 22–30 (2010).
    1. Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav. Immun. 26(8), 1191–1201 (2012).
    1. Maier B, Laurer H-L, Rose S, Buurman WA, Marzi I. Physiological levels of pro- and anti-inflammatory mediators in cerebrospinal fluid and plasma: a normative study. J. Neurotrauma. 22(7), 822–835 (2005).
    1. Chiaretti A, Genovese O, Aloe L, et al. Interleukin 1? and interleukin 6 relationship with paediatric head trauma severity and outcome. Childs Nerv Syst. 21(3), 185–193 (2004).
    1. Folkersma H, Brevé JJP, Tilders FJH, Cherian L, Robertson CS, Vandertop WP. Cerebral microdialysis of interleukin (IL)-1beta and IL-6: extraction efficiency and production in the acute phase after severe traumatic brain injury in rats. Acta Neurochir. 150(12), 1277–1284 (2008).
    1. Bonneh-Barkay D, Zagadailov P, Zou H, et al. YKL-40 expression in traumatic brain injury: an initial analysis. J. Neurotrauma. 27(7), 1215–1223 (2010).
    1. Kang J-H, Lin H-C. Increased risk of multiple sclerosis after traumatic brain injury: a nationwide population-based study. J. Neurotrauma. 29(1), 90–95 (2012).
    1. Dams-O’Connor K, Gibbons LE, Bowen JD, McCurry SM, Larson EB, Crane PK. Risk for late-life re-injury, dementia and death among individuals with traumatic brain injury: a population-based study. J. Neurol. Neurosurg. Psychiatr. 84(2), 177–182 (2013).
    1. Sivanandam TM, Thakur MK. Traumatic brain injury: a risk factor for Alzheimer’s disease. Neurosci Biobehav Rev. 36(5), 1376–1381 (2012).
    1. Lee P-C, Bordelon Y, Bronstein J, Ritz B. Traumatic brain injury, paraquat exposure, and their relationship to Parkinson disease. Neurology. 79(20), 2061–2066 (2012).
    1. Smith DH, Chen XH, Nonaka M, et al. Accumulation of amyloid beta and tau and the formation of neurofilament inclusions following diffuse brain injury in the pig. 58(9), 982–992 (1999).
    1. McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. 68(7), 709–735 (2009).
    1. McKee AC, Stern RA, Nowinski CJ, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain. 136(Pt 1), 43–64 (2013).

      ** A key review paper describing post-TBI chronic traumatic encephalopathy.

    1. Omalu BI, Hamilton RL, Kamboh MI, DeKosky ST, Bailes J. Chronic traumatic encephalopathy (CTE) in a National Football League Player: Case report and emerging medicolegal practice questions. Journal of Forensic Nursing. 6(1), 40–46 (2010).
    1. Duan Y, Dong S, Gu F, Hu Y, Zhao Z. Advances in the Pathogenesis of Alzheimer’s Disease: Focusing on Tau-Mediated Neurodegeneration. Transl Neurodegener. 1(1), 24 (2012).
    1. Yang Z, Wang P, Morgan D, et al. Temporal MRI characterization, neurobiochemical and neurobehavioral changes in a mouse repetitive concussive head injury model. Sci. Rep. 5(1), 11178 (2015).
    1. 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. J. Neurotrauma. 32(5), 342–352 (2015).
    1. Goldstein LE, Fisher AM, Tagge CA, et al. Chronic Traumatic Encephalopathy in Blast-Exposed Military Veterans and a Blast Neurotrauma Mouse Model. Science Translational Medicine [Internet]. 4(134), 134ra60–134ra60 (2012).
    1. Shahim P, Tegner Y, Wilson DH, et al. Blood biomarkers for brain injury in concussed professional ice hockey players. JAMA Neurol. 71(6), 684–692 (2014).

      * First paper describing the detection of Tau elevations in blood in the acute phase following concussions, with the use of high sensitivity assay platform (SIMOA).

    1. Olivera A, Lejbman N, Jeromin A, et al. Peripheral Total Tau in Military Personnel Who Sustain Traumatic Brain Injuries During Deployment. JAMA Neurol. 72(10), 1109–1116 (2015).
    1. 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 Neurol. 74(9), 1063–1072 (2017).

      * First paper to show P-Tau (and Tau) elevations in blood not only in the acute phase but also chronic phase of TBI, using a high sensitivity platform (SOFIA).

    1. Yang Z, Lin F, Robertson CS, Wang KKW. Dual vulnerability of TDP-43 to calpain and caspase-3 proteolysis after neurotoxic conditions and traumatic brain injury. Journal of Cerebral Blood Flow & Metabolism. 34(9), 1444–1452 (2014).
    1. Dambinova SAS, Khounteev GAG, Izykenova GAG, Zavolokov IGI, Ilyukhina AYA, Skoromets AAA. Blood test detecting autoantibodies to N-methyl-D-aspartate neuroreceptors for evaluation of patients with transient ischemic attack and stroke. Clinical Chemistry. 49(10), 1752–1762 (2003).
    1. Colasanti TT, Barbati CC, Rosano GG, Malorni WW, Ortona EE. Autoantibodies in patients with Alzheimer’s disease: pathogenetic role and potential use as biomarkers of disease progression. Autoimmunity Reviews. 9(12), 807–811 (2010).
    1. D’Andrea MR. Add Alzheimer’s disease to the list of autoimmune diseases. Medical Hypotheses. 64(3), 458–463 (2005).
    1. Ankeny DP, Lucin KM, Sanders VM, McGaughy VM, Popovich PG. Spinal cord injury triggers systemic autoimmunity: evidence for chronic B lymphocyte activation and lupus-like autoantibody synthesis. J. Neurochem. 99(4), 1073–1087 (2006).
    1. Popovich PGP, Stokes BTB, Whitacre CCC. Concept of autoimmunity following spinal cord injury: possible roles for T lymphocytes in the traumatized central nervous system. J. Neurosci. Res. 45(4), 349–363 (1996).
    1. Lancaster E, Dalmau J. Neuronal autoantigens—pathogenesis, associated disorders and antibody testing. Nature Reviews Neurology. 8(7), 380–390 (2012).
    1. Ponomarenko NA, Durova OM, Vorobiev II, et al. Catalytic activity of autoantibodies toward myelin basic protein correlates with the scores on the multiple sclerosis expanded disability status scale. Immunology Letters. 103(1), 45–50 (2006).
    1. Hedegaard CJ, Chen N, Sellebjerg F, et al. Autoantibodies to myelin basic protein (MBP) in healthy individuals and in patients with multiple sclerosis: a role in regulating cytokine responses to MBP. Immunology [Internet]. 128(1pt2), e451–e461 (2009).
    1. Cox AL, Coles AJ, Nortje J, et al. An investigation of auto-reactivity after head injury. Journal of Neuroimmunology. 174(1–2), 180–186 (2006).
    1. Goryunova AVA, Bazarnaya NAN, Sorokina EGE, et al. Glutamate receptor autoantibody concentrations in children with chronic post-traumatic headache. Neurosci Behav Physiol. 37(8), 761–764 (2007).
    1. Marchi N, Bazarian JJ, Puvenna V, et al. Consequences of Repeated Blood-Brain Barrier Disruption in Football Players. PLoS ONE. 8(3), e56805 (2013).
    1. Wang KKW, Yang Z, Yue JK, et al. Plasma Anti-Glial Fibrillary Acidic Protein Autoantibody Levels during the Acute and Chronic Phases of Traumatic Brain Injury: A Transforming Research and Clinical Knowledge in Traumatic Brain Injury Pilot Study. J. Neurotrauma. 33(13), 1270–1277 (2016).
    1. Tanriverdi F, De Bellis A, Bizzarro A, et al. Antipituitary antibodies after traumatic brain injury: is head trauma-induced pituitary dysfunction associated with autoimmunity? European Journal of Endocrinology. 159(1), 7–13 (2008).
    1. Tanriverdi F, De Bellis A, Battaglia M, et al. Investigation of antihypothalamus and antipituitary antibodies in amateur boxers: is chronic repetitive head trauma-induced pituitary dysfunction associated with autoimmunity? European Journal of Endocrinology. 162(5), 861–867 (2010).
    1. Aimaretti G, Ambrosio MR, Di Somma C, et al. Hypopituitarism induced by traumatic brain injury in the transition phase. J. Endocrinol. Invest. 28(11), 984–989 (2005).
    1. Tanriverdi F, De Bellis A, Ulutabanca H, et al. A Five Year Prospective Investigation of Anterior Pituitary Function after Traumatic Brain Injury: Is Hypopituitarism Long-Term after Head Trauma Associated with Autoimmunity? J. Neurotrauma. 30(16), 1426–1433 (2013).
    1. Yang Z, Zhu T, Weissman AS, et al. Autoimmunity and Traumatic Brain Injury. 1–8 (2017).
    1. Mondello S, Gabrielli A, Catani 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(13–14), 1629–1635 (2012).
    1. Papa L, Robicsek SA, Brophy GM, et al. Temporal Profile of Microtubule-Associated Protein 2: A Novel Indicator of Diffuse Brain Injury Severity and Early Mortality after Brain Trauma. J. Neurotrauma. neu.20174994–9 (2017).
    1. Korley FK, Diaz-Arrastia R, Wu AHB, et al. Circulating Brain-Derived Neurotrophic Factor Has Diagnostic and Prognostic Value in Traumatic Brain Injury. J. Neurotrauma. 33(2), 215–225 (2016).
    1. Yang J, Korley FK, Dai M, Everett AD. Serum neurogranin measurement as a biomarker of acute traumatic brain injury. Clin. Biochem. 48(13–14), 843–848 (2015).
    1. Kenney K, Amyot F, Haber M, et al. Cerebral Vascular Injury in Traumatic Brain Injury. Exp. Neurol. 275(Part 3), 353–366 (2016).
    1. Tomkins O, Feintuch A, Benifla M, Cohen A, Friedman A, Shelef I. Blood-Brain Barrier Breakdown Following Traumatic Brain Injury: A Possible Role in Posttraumatic Epilepsy. Cardiovascular Psychiatry and Neurology. 765923, 1–11 (2011).
    1. Muradashvili N, Lominadze D. Role of fibrinogen in cerebrovascular dysfunction after traumatic brain injury. Brain Inj. 27(13–14), 1508–1515 (2013).
    1. De Oliveira CO, Reimer AG, Da Rocha AB, et al. Plasma von Willebrand factor levels correlate with clinical outcome of severe traumatic brain injury. J. Neurotrauma. 24(8), 1331–1338 (2007).
    1. Berger RP, Fromkin J, Rubin P, Snyder J, Richichi R, Kochanek P. Serum D-dimer concentrations are increased after pediatric traumatic brain injury. J. Pediatr. 166(2), 383–388 (2015).
    1. Balakathiresan N, Bhomia M, Chandran R, Chavko M, McCarron RM, Maheshwari RK. MicroRNA Let-7i Is a Promising Serum Biomarker for Blast-Induced Traumatic Brain Injury. J. Neurotrauma. 29(7), 1379–1387 (2012).
    1. Redell JB, Moore AN, Ward NH, Hergenroeder GW, Dash PK. Human traumatic brain injury alters plasma microRNA levels. J. Neurotrauma. 27(12), 2147–2156 (2010).
    1. Bhomia M, Balakathiresan NS, Wang KK, Papa L, Maheshwari RK. A Panel of Serum MiRNA Biomarkers for the Diagnosis of Severe to Mild Traumatic Brain Injury in Humans. Sci. Rep. 6(1), 28148–12 (2016).

      * A comprehensive study identifying a panel of CSF- and blood-based miRNA’s as candidate biomarkers for mild to severe TBI in human.

    1. Mitra B, Rau TF, Surendran N, 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).
    1. Di Pietro V, Ragusa M, Davies D, et al. MicroRNAs as Novel Biomarkers for the Diagnosis and Prognosis of Mild and Severe Traumatic Brain Injury. J. Neurotrauma. 34(11), 1948–1956 (2017).
    1. Ferrari M, Cremonesi L, Galbiati S. 10. Circulating Nucleic Acids as Diagnostic Tool. EJIFCC. 19(1), 68–74 (2008).
    1. Rodrigues Filho EM, Simon D, Ikuta N, et al. Elevated cell-free plasma DNA level as an independent predictor of mortality in patients with severe traumatic brain injury. J. Neurotrauma. 31(19), 1639–1646 (2014).
    1. Campello Yurgel V, Ikuta N, Brondani da Rocha A, et al. Role of Plasma DNA as a Predictive Marker of Fatal Outcome following Severe Head Injury in Males. J. Neurotrauma. 24(7), 1172–1181 (2007).
    1. Macher H, Egea-Guerrero JJ, Revuelto-Rey J, et al. Role of early cell-free DNA levels decrease as a predictive marker of fatal outcome after severe traumatic brain injury. Clinica Chimica Acta. 414(C), 12–17 (2012).
    1. Rodrigues Filho EM, Ikuta N, Simon D, Regner AP. Prognostic value of circulating DNA levels in critically ill and trauma patients. Revista Brasileira de Terapia Intensiva. 26(3), 1–8 (2014).
    1. Agoston DV, Shutes-David A, Peskind ER. Biofluid biomarkers of traumatic brain injury. Brain Injury. 31(9), 1195–1203 (2017).
    1. Moyron RB, Wall NR. Differential protein expression in exosomal samples taken from trauma patients. Proteomics Clin Appl. 11(9–10), 1700095 (2017).
    1. Manek R, Moghieb A, Yang Z, et al. Protein Biomarkers and Neuroproteomics Characterization of Microvesicles/Exosomes from Human Cerebrospinal Fluid Following Traumatic Brain Injury. Mol Neurobiol. 24(2), 133–31 (2017).
    1. Nekludov M, Bellander BM, Gryth D, Wallen H, Mobarrez F. Brain-Derived Microparticles in Patients with Severe Isolated TBI. Brain Injury. 31(13–14), 1856–1862 (2017).
    1. Stern RA, Tripodis Y, Baugh CM, et al. Preliminary Study of Plasma Exosomal Tau as a Potential Biomarker for Chronic Traumatic Encephalopathy. J Alzheimers Dis. 51(4), 1099–1109 (2016).
    1. McCrory P, Davis G, Makdissi M. Second impact syndrome or cerebral swelling after sporting head injury. Curr Sports Med Rep. 11(1), 21–23 (2012).
    1. Cantu RC. Second Impact Syndrome: Concussion and Second Injury Brain Complications. Yearbook of Sports Medicine. 2011, 26–28 (2011).
    1. Stovitz SD, Weseman JD, Hooks MC, Schmidt RJ, Koffel JB, Patricios JS. What Definition Is Used to Describe Second Impact Syndrome in Sports? A Systematic and Critical Review. Curr Sports Med Rep. 16(1), 50–55 (2017).
    1. Thelin EP, Nelson DW, Bellander B-M. Secondary Peaks of S100B in Serum Relate to Subsequent Radiological Pathology in Traumatic Brain Injury. Neurocrit Care. 20(2), 217–229 (2013).
    1. Kellermann I, Kleindienst A, Hore N, Buchfelder M, Brandner S. Early CSF and Serum S100B Concentrations for Outcome Prediction in Traumatic Brain Injury and Subarachnoid Hemorrhage. Clin Neurol Neurosurg. 145, 79–83 (2016).
    1. Di Battista AP, Buonora JE, Rhind SG, et al. Blood Biomarkers in Moderate-To-Severe Traumatic Brain Injury: Potential Utility of a Multi-Marker Approach in Characterizing Outcome. Front Neurol. 6(8), 45–9 (2015).
    1. Egea-Guerrero JJ, Murillo-Cabezas F, Gordillo-Escobar E, et al. S100B protein may detect brain death development after severe traumatic brain injury. J. Neurotrauma. 30(20), 1762–1769 (2013).
    1. Rainey T, Lesko M, Sacho R, Lecky F, Childs C. Predicting outcome after severe traumatic brain injury using the serum S100B biomarker: results using a single (24h) time-point. Resuscitation. 80(3), 341–345 (2009).
    1. Lee JY, Lee CY, Kim HR, Lee C-H, Kim HW, Kim JH. A Role of Serum-Based Neuronal and Glial Markers as Potential Predictors for Distinguishing Severity and Related Outcomes in Traumatic Brain Injury. J Korean Neurosurg Soc. 58(2), 93–100 (2015).
    1. Thelin EP, Nelson DW, Bellander B-M. A review of the clinical utility of serum S100B protein levels in the assessment of traumatic brain injury. Acta Neurochir. 159(2), 209–225 (2017).
    1. Pelinka LE, Kroepfl A, Leixnering M, Buchinger W, Raabe A, Redl H. GFAP Versus S100B in Serum after Traumatic Brain Injury: Relationship to Brain Damage and Outcome. J. Neurotrauma. 21(11), 1553–1561 (2004).
    1. Lei J, Gao G, Feng J, et al. Glial fibrillary acidic protein as a biomarker in severe traumatic brain injury patients: a prospective cohort study. Critical care (London, England). 19(1), 362–12 (2015).
    1. Czeiter E, Mondello S, Kovacs N, et al. Brain injury biomarkers may improve the predictive power of the IMPACT outcome calculator. J. Neurotrauma. 29(9), 1770–1778 (2012).
    1. Mondello S, Kobeissy F, Vestri A, Hayes RL, Kochanek PM, Berger RP. Serum Concentrations of Ubiquitin C-Terminal Hydrolase-L1 and Glial Fibrillary Acidic Protein after Pediatric Traumatic Brain Injury. Sci. Rep. 6, 28203 (2016).
    1. Fraser DD, Close TE, Rose KL, et al. Severe traumatic brain injury in children elevates glial fibrillary acidic protein in cerebrospinal fluid and serum*. Pediatr Crit Care Med. 12(3), 319–324 (2011).
    1. Papa L, Lewis LM, Silvestri S, et al. Serum levels of ubiquitin C-terminal hydrolase distinguish mild traumatic brain injury from trauma controls and are elevated in mild and moderate traumatic brain injury patients with intracranial lesions and neurosurgical intervention. J Trauma Acute Care Surg. 72(5), 1335–1344 (2012).
    1. Takala RSK, Posti JP, Runtti H, et al. Glial Fibrillary Acidic Protein and Ubiquitin C-Terminal Hydrolase-L1 as Outcome Predictors in Traumatic Brain Injury. World Neurosurgery. 87, 8–20 (2016).
    1. Shahim P, Gren M, Liman V, et al. Serum neurofilament light protein predicts clinical outcome in traumatic brain injury. Sci. Rep. 6, 36791 (2016).
    1. Žurek J, Fedora M. The usefulness of S100B, NSE, GFAP, NF-H, secretagogin and Hsp70 as a predictive biomarker of outcome in children with traumatic brain injury. Acta Neurochir. 154(1), 93–103– discussion 103 (2011).
    1. Liliang P-C, Liang C-L, Lu K, et al. Relationship between injury severity and serum tau protein levels in traumatic brain injured rats. Resuscitation. 81(9), 1205–1208 (2010).
    1. Zemlan FP, Jauch EC, Mulchahey JJ, et al. C-tau biomarker of neuronal damage in severe brain injured patients: association with elevated intracranial pressure and clinical outcome. Brain research. 947(1), 131–139 (2002).
    1. Pandey S, Singh K, Sharma V, et al. A prospective pilot study on serum cleaved tau protein as a neurological marker in severe traumatic brain injury. Br J Neurosurg. 73, 1–8 (2017).
    1. 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. J. Neurotrauma. 34(1), 66–73 (2017).
    1. Abbasi M, Sajjadi M, Fathi M, Maghsoudi M. Serum S100B Protein as an Outcome Prediction Tool in Emergency Department Patients with Traumatic Brain Injury. Turk J Emerg Med. 14(4), 147–152 (2014).
    1. 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 Inj. 20(7), 759–765 (2006).
    1. Kochanek PM, Bramlett HM, Dixon CE, et al. Operation Brain Trauma Therapy: Approach to Modeling, Therapy Evaluation, Drug Selection, and Biomarker Assessments, for a Multi-Center Pre-Clinical Drug Screening Consortium for Acute Therapies in Severe Traumatic Brain Injury. J. Neurotrauma. neu.20154113 (2015).
    1. Kochanek PM, Bramlett H, Dietrich WD, et al. A novel multicenter preclinical drug screening and biomarker consortium for experimental traumatic brain injury: operation brain trauma therapy. J Trauma. 71(1 Suppl), S15–S24 (2011).
    1. Mondello S, Shear DA, Bramlett HM, et al. Insight into Preclinical Models of Traumatic Brain Injury Using Circulating Brain Damage Biomarkers: Operation Brain Trauma Therapy. J. Neurotrauma. neu.20154132 (2015).
    1. Mountney A, Bramlett HM, Dixon CE, et al. Simvastatin Treatment in Traumatic Brain Injury: Operation Brain Trauma Therapy. J. Neurotrauma [Internet]. neu.20154130 (2015).
    1. Shear DA, Bramlett HM, Dixon CE, et al. Nicotinamide Treatment in Traumatic Brain Injury: Operation Brain Trauma Therapy. J. Neurotrauma. neu.20154115 (2015).
    1. Wei HH, Lu XC, Shear DA, et al. NNZ-2566 treatment inhibits neuroinflammation and pro-inflammatory cytokine expression induced by experimental penetrating ballistic-like brain injury in rats. Journal of Neuroinflammation. 6, 19 (2009).
    1. Wright DW, Kellermann AL, Hertzberg VS, et al. ProTECT: A Randomized Clinical Trial of Progesterone for Acute Traumatic Brain Injury. Ann Emerg Med. 49(4), 391–402.e2 (2007).
    1. Wright DW, Yeatts SD, Silbergleit R, et al. Very early administration of progesterone for acute traumatic brain injury. N Engl J Med. 371(26), 2457–2466 (2014).
    1. Kobeissy FH, Guingab-Cagmat JD, Razafsha M, et al. Leveraging biomarker platforms and systems biology for rehabilomics and biologics effectiveness research. PM & R. 3(6, Supplement), S139–S147 (2011).
    1. Zhang Z, Mondello S, Kobeissy F, et al. Protein biomarkers for traumatic and ischemic brain injury: from bench to bedside. Translational Stroke Research [Internet]. 3, 1–32 (2011).
    1. Varma S, Janesko KL, Wisniewski SR, et al. F2-isoprostane and neuron-specific enolase in cerebrospinal fluid after severe traumatic brain injury in infants and children. J. Neurotrauma. 20(8), 781–786 (2003).
    1. Berger RP, Kochanek PM, Pierce MC. Biochemical markers of brain injury: could they be used as diagnostic adjuncts in cases of inflicted traumatic brain injury? Child Abuse & Neglect. 28(7), 739–754 (2004).
    1. Wagner AK, Bayir H, Ren D, Puccio A, Zafonte RD, Kochanek PM. Relationships between cerebrospinal fluid markers of excitotoxicity, ischemia, and oxidative damage after severe TBI: the impact of gender, age, and hypothermia. J. Neurotrauma. 21(2), 125–136 (2004).

Source: PubMed

3
Se inscrever