Semin Neurol 2020; 40(04): 430-438
DOI: 10.1055/s-0040-1713620
Review Article

Repetitive Head Trauma Induces Chronic Traumatic Encephalopathy by Multiple Mechanisms

Jonathan D. Cherry
1   Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts
2   Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
3   Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
4   VA Boston Healthcare System, Boston, Massachusetts
,
Katharine J. Babcock
1   Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts
4   VA Boston Healthcare System, Boston, Massachusetts
5   Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts
6   Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts
,
Lee E. Goldstein
1   Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts
3   Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
5   Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts
7   Boston University College of Engineering, Boston University, Boston, Massachusetts
› Institutsangaben

Abstract

Exposure to repetitive neurotrauma increases lifetime risk for developing progressive cognitive deficits, neurobehavioral abnormalities, and chronic traumatic encephalopathy (CTE). CTE is a tau protein neurodegenerative disease first identified in boxers and recently described in athletes participating in other contact sports (notably American football, ice hockey, rugby, and wrestling) and in military veterans with blast exposure. Currently, CTE can only be diagnosed by neuropathological examination of the brain after death. The defining diagnostic lesion of CTE consists of patchy perivascular accumulations of hyperphosphorylated tau protein that localize in the sulcal depths of the cerebral cortex. Neuronal abnormalities, axonopathy, neurovascular dysfunction, and neuroinflammation are triggered by repetitive head impacts (RHIs) and likely act as catalysts for CTE pathogenesis and progression. However, the specific mechanisms that link RHI to CTE are unknown. This review will explore two important areas of CTE pathobiology. First, we will review what is known about the biomechanical properties of RHI that initiate CTE-related pathologies. Second, we will provide an overview of key features of CTE neuropathology and how these contribute to abnormal tau hyperphosphorylation, accumulation, and spread.



Publikationsverlauf

Artikel online veröffentlicht:
16. Juli 2020

© 2020. Thieme. All rights reserved.

Thieme Medical Publishers
333 Seventh Avenue, New York, NY 10001, USA.

 
  • References

  • 1 Surveillance Report of Traumatic Brain Injury-related Emergency Department Visits, Hospitalizations, and Deaths—United States, 2014. Centers for Disease Control and Prevention, U.S. Department of Health and Human Services; 2019
  • 2 Lumba-Brown A, Yeates KO, Sarmiento K. , et al. Centers for Disease Control and Prevention guideline on the diagnosis and management of mild traumatic brain injury among children. JAMA Pediatr 2018; 172 (11) e182853
  • 3 Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact of traumatic brain injury: a brief overview. J Head Trauma Rehabil 2006; 21 (05) 375-378
  • 4 Wojnarowicz MW, Fisher AM, Minaeva O, Goldstein LE. Considerations for experimental animal models of concussion, traumatic brain injury, and chronic traumatic encephalopathy-these matters matter. Front Neurol 2017; 8: 240
  • 5 Shahim P, Tegner Y, Gustafsson B. , et al. Neurochemical aftermath of repetitive mild traumatic brain injury. JAMA Neurol 2016; 73 (11) 1308-1315
  • 6 McKee AC, Alosco ML, Huber BR. Repetitive head impacts and chronic traumatic encephalopathy. Neurosurg Clin N Am 2016; 27 (04) 529-535
  • 7 Roberts AL, Pascual-Leone A, Speizer FE. , et al. Exposure to American football and neuropsychiatric health in former National Football League players: findings from the football players health study. Am J Sports Med 2019; 47 (12) 2871-2880
  • 8 Alosco ML, Mez J, Tripodis Y. , et al. Age of first exposure to tackle football and chronic traumatic encephalopathy. Ann Neurol 2018; 83 (05) 886-901
  • 9 Mez J, Daneshvar DH, Kiernan PT. , et al. Clinicopathological evaluation of chronic traumatic encephalopathy in players of American football. JAMA 2017; 318 (04) 360-370
  • 10 McKee AC, Cairns NJ, Dickson DW. , et al; TBI/CTE group. The first NINDS/NIBIB consensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol 2016; 131 (01) 75-86
  • 11 Cherry JD, Tripodis Y, Alvarez VE. , et al. Microglial neuroinflammation contributes to tau accumulation in chronic traumatic encephalopathy. Acta Neuropathol Commun 2016; 4 (01) 112
  • 12 Tagge CA, Fisher AM, Minaeva OV. , et al. Concussion, microvascular injury, and early tauopathy in young athletes after impact head injury and an impact concussion mouse model. Brain 2018; 141 (02) 422-458
  • 13 Goldstein LE, Fisher AM, Tagge CA. , et al. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci Transl Med 2012; 4 (134) 134ra60
  • 14 McKee AC, Daneshvar DH, Alvarez VE, Stein TD. The neuropathology of sport. Acta Neuropathol 2014; 127 (01) 29-51
  • 15 Mez J, Solomon TM, Daneshvar DH. , et al. Assessing clinicopathological correlation in chronic traumatic encephalopathy: rationale and methods for the UNITE study. Alzheimers Res Ther 2015; 7 (01) 62
  • 16 Kondo A, Shahpasand K, Mannix R. , et al. Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature 2015; 523 (7561): 431-436
  • 17 Faden AI, Demediuk P, Panter SS, Vink R. The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science 1989; 244 (4906): 798-800
  • 18 Katayama Y, Becker DP, Tamura T, Hovda DA. Massive increases in extracellular potassium and the indiscriminate release of glutamate following concussive brain injury. J Neurosurg 1990; 73 (06) 889-900
  • 19 Giza CC, Hovda DA. The new neurometabolic cascade of concussion. Neurosurgery 2014; 75 (Suppl. 04) S24-S33
  • 20 Barkhoudarian G, Hovda DA, Giza CC. The molecular pathophysiology of concussive brain injury - an update. Phys Med Rehabil Clin N Am 2016; 27 (02) 373-393
  • 21 Bieniek KF, Ross OA, Cormier KA. , et al. Chronic traumatic encephalopathy pathology in a neurodegenerative disorders brain bank. Acta Neuropathol 2015; 130 (06) 877-889
  • 22 Ghajari M, Hellyer PJ, Sharp DJ. Computational modelling of traumatic brain injury predicts the location of chronic traumatic encephalopathy pathology. Brain 2017; 140 (02) 333-343
  • 23 Cloots RJ, Gervaise HM, van Dommelen JA, Geers MG. Biomechanics of traumatic brain injury: influences of the morphologic heterogeneities of the cerebral cortex. Ann Biomed Eng 2008; 36 (07) 1203-1215
  • 24 Shreiber DI, Smith DH, Meaney DF. Immediate in vivo response of the cortex and the blood-brain barrier following dynamic cortical deformation in the rat. Neurosci Lett 1999; 259 (01) 5-8
  • 25 Huber BR, Meabon JS, Martin TJ. , et al. Blast exposure causes early and persistent aberrant phospho- and cleaved-tau expression in a murine model of mild blast-induced traumatic brain injury. J Alzheimers Dis 2013; 37 (02) 309-323
  • 26 Weissberg I, Wood L, Kamintsky L. , et al. Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction. Neurobiol Dis 2015; 78: 115-125
  • 27 Smith DH, Meaney DF, Shull WH. Diffuse axonal injury in head trauma. J Head Trauma Rehabil 2003; 18 (04) 307-316
  • 28 Hemphill MA, Dauth S, Yu CJ, Dabiri BE, Parker KK. Traumatic brain injury and the neuronal microenvironment: a potential role for neuropathological mechanotransduction. Neuron 2015; 85 (06) 1177-1192
  • 29 Annegers JF, Hauser WA, Coan SP, Rocca WA. A population-based study of seizures after traumatic brain injuries. N Engl J Med 1998; 338 (01) 20-24
  • 30 McKee AC, Stein TD, Kiernan PT, Alvarez VE. The neuropathology of chronic traumatic encephalopathy. Brain Pathol 2015; 25 (03) 350-364
  • 31 Graham DI, Gennarelli TA, McIntosh TK. Greenfield's Neuropathology. In: Graham DL. , ed. London: Arnold; 2002: 823-898
  • 32 Greenfield J, Love S, Louis D. Greenfield's Neuropathology. 8th edn. London: Hodder Arnold; 2008
  • 33 Smith C, Gentleman SM, Leclercq PD. , et al. The neuroinflammatory response in humans after traumatic brain injury. Neuropathol Appl Neurobiol 2013; 39 (06) 654-666
  • 34 Marion CM, Radomski KL, Cramer NP, Galdzicki Z, Armstrong RC. Experimental traumatic brain injury identifies distinct early and late phase axonal conduction deficits of white matter pathophysiology, and reveals intervening recovery. J Neurosci 2018; 38 (41) 8723-8736
  • 35 Smith DH, Wolf JA, Lusardi TA, Lee VM, Meaney DF. High tolerance and delayed elastic response of cultured axons to dynamic stretch injury. J Neurosci 1999; 19 (11) 4263-4269
  • 36 Johnson VE, Stewart W, Smith DH. Axonal pathology in traumatic brain injury. Exp Neurol 2013; 246: 35-43
  • 37 Holleran L, Kim JH, Gangolli M. , et al. Axonal disruption in white matter underlying cortical sulcus tau pathology in chronic traumatic encephalopathy. Acta Neuropathol 2017; 133 (03) 367-380
  • 38 Shively SB, Edgerton SL, Iacono D. , et al. Localized cortical chronic traumatic encephalopathy pathology after single, severe axonal injury in human brain. Acta Neuropathol 2017; 133 (03) 353-366
  • 39 Sweeney MD, Kisler K, Montagne A, Toga AW, Zlokovic BV. The role of brain vasculature in neurodegenerative disorders. Nat Neurosci 2018; 21 (10) 1318-1331
  • 40 Attwell D, Buchan AM, Charpak S, Lauritzen M, Macvicar BA, Newman EA. Glial and neuronal control of brain blood flow. Nature 2010; 468 (7321): 232-243
  • 41 Parri R, Crunelli V. An astrocyte bridge from synapse to blood flow. Nat Neurosci 2003; 6 (01) 5-6
  • 42 Nedergaard M. Neuroscience. Garbage truck of the brain. Science 2013; 340 (6140): 1529-1530
  • 43 Simon DW, McGeachy MJ, Bayır H, Clark RS, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol 2017; 13 (03) 171-191
  • 44 Iliff JJ, Chen MJ, Plog BA. , et al. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J Neurosci 2014; 34 (49) 16180-16193
  • 45 Iliff JJ, Wang M, Liao Y. , et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 2012; 4 (147) 147ra111
  • 46 Karve IP, Taylor JM, Crack PJ. The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol 2016; 173 (04) 692-702
  • 47 Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci 2006; 7 (01) 41-53
  • 48 Iadecola C. Neurovascular regulation in the normal brain and in Alzheimer's disease. Nat Rev Neurosci 2004; 5 (05) 347-360
  • 49 Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 2008; 57 (02) 178-201
  • 50 Argaw AT, Gurfein BT, Zhang Y, Zameer A, John GR. VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown. Proc Natl Acad Sci U S A 2009; 106 (06) 1977-1982
  • 51 Cherry JD, Stein TD, Tripodis Y. , et al. CCL11 is increased in the CNS in chronic traumatic encephalopathy but not in Alzheimer's disease. PLoS One 2017; 12 (09) e0185541
  • 52 Cherry JD, Mez J, Crary JF. , et al. Variation in TMEM106B in chronic traumatic encephalopathy. Acta Neuropathol Commun 2018; 6 (01) 115
  • 53 Cherry JD, Zeineddin A, Dammer EB. , et al. Characterization of detergent insoluble proteome in chronic traumatic encephalopathy. J Neuropathol Exp Neurol 2018; 77 (01) 40-49
  • 54 Coughlin JM, Wang Y, Munro CA. , et al. Neuroinflammation and brain atrophy in former NFL players: an in vivo multimodal imaging pilot study. Neurobiol Dis 2015; 74: 58-65
  • 55 Coughlin JM, Wang Y, Minn I. , et al. Imaging of glial cell activation and white matter integrity in brains of active and recently retired National Football League players. JAMA Neurol 2017; 74 (01) 67-74
  • 56 Cherry JD, Olschowka JA, O'Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation 2014; 11 (01) 98
  • 57 Carmona S, Zahs K, Wu E, Dakin K, Bras J, Guerreiro R. The role of TREM2 in Alzheimer's disease and other neurodegenerative disorders. Lancet Neurol 2018; 17 (08) 721-730
  • 58 Ulrich JD, Ulland TK, Colonna M, Holtzman DM. Elucidating the role of TREM2 in Alzheimer's disease. Neuron 2017; 94 (02) 237-248
  • 59 Correa FG, Hernangómez M, Guaza C. Understanding microglia-neuron cross talk: relevance of the microglia-neuron cocultures. Methods Mol Biol 2013; 1041: 215-229
  • 60 Jurgens HA, Johnson RW. Dysregulated neuronal-microglial cross-talk during aging, stress and inflammation. Exp Neurol 2012; 233 (01) 40-48
  • 61 Puntambekar SS, Saber M, Lamb BT, Kokiko-Cochran ON. Cellular players that shape evolving pathology and neurodegeneration following traumatic brain injury. Brain Behav Immun 2018; 71: 9-17
  • 62 Schulz C, Gomez Perdiguero E, Chorro L. , et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012; 336 (6077): 86-90
  • 63 Mckee AC, Daneshvar DH. The neuropathology of traumatic brain injury. Handb Clin Neurol 2015; 127: 45-66
  • 64 Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci U S A 1975; 72 (05) 1858-1862
  • 65 Goedert M, Eisenberg DS, Crowther RA. Propagation of Tau aggregates and neurodegeneration. Annu Rev Neurosci 2017; 40: 189-210
  • 66 Kopeikina KJ, Carlson GA, Pitstick R. , et al. Tau accumulation causes mitochondrial distribution deficits in neurons in a mouse model of tauopathy and in human Alzheimer's disease brain. Am J Pathol 2011; 179 (04) 2071-2082
  • 67 Hoover BR, Reed MN, Su J. , et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 2010; 68 (06) 1067-1081
  • 68 Chong FP, Ng KY, Koh RY, Chye SM. Tau proteins and tauopathies in Alzheimer's disease. Cell Mol Neurobiol 2018; 38 (05) 965-980
  • 69 Šimić G, Babić Leko M, Wray S. , et al. Tau protein hyperphosphorylation and aggregation in Alzheimer's disease and other tauopathies, and possible neuroprotective strategies. Biomolecules 2016; 6 (01) 6
  • 70 Lu KP, Kondo A, Albayram O, Herbert MK, Liu H, Zhou XZ. Potential of the antibody against cis-phosphorylated tau in the early diagnosis, treatment, and prevention of Alzheimer disease and brain injury. JAMA Neurol 2016; 73 (11) 1356-1362
  • 71 McKee AC, Stern RA, Nowinski CJ. , et al. The spectrum of disease in chronic traumatic encephalopathy. Brain 2013; 136 (Pt 1): 43-64
  • 72 Spires-Jones TL, Hyman BT. The intersection of amyloid beta and tau at synapses in Alzheimer's disease. Neuron 2014; 82 (04) 756-771
  • 73 Goedert M, Masuda-Suzukake M, Falcon B. Like prions: the propagation of aggregated tau and α-synuclein in neurodegeneration. Brain 2017; 140 (02) 266-278
  • 74 de Calignon A, Polydoro M, Suárez-Calvet M. , et al. Propagation of tau pathology in a model of early Alzheimer's disease. Neuron 2012; 73 (04) 685-697
  • 75 Montenigro PH, Corp DT, Stein TD, Cantu RC, Stern RA. Chronic traumatic encephalopathy: historical origins and current perspective. Annu Rev Clin Psychol 2015; 11 (01) 309-330
  • 76 Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem 2009; 284 (19) 12845-12852
  • 77 Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer's-like tauopathy. J Neurosci 2013; 33 (03) 1024-1037
  • 78 Wu JW, Herman M, Liu L. , et al. Small misfolded Tau species are internalized via bulk endocytosis and anterogradely and retrogradely transported in neurons. J Biol Chem 2013; 288 (03) 1856-1870
  • 79 Asai H, Ikezu S, Tsunoda S. , et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 2015; 18 (11) 1584-1593
  • 80 Stopschinski BE, Diamond MI. The prion model for progression and diversity of neurodegenerative diseases. Lancet Neurol 2017; 16 (04) 323-332
  • 81 Sanders DW, Kaufman SK, DeVos SL. , et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 2014; 82 (06) 1271-1288
  • 82 Zanier ER, Bertani I, Sammali E. , et al. Induction of a transmissible tau pathology by traumatic brain injury. Brain 2018; 141 (09) 2685-2699
  • 83 Zilka N, Kazmerova Z, Jadhav S. , et al. Who fans the flames of Alzheimer's disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation 2012; 9: 47
  • 84 Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer's disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov 2011; 10 (09) 698-712
  • 85 Hardy JA, Higgins GA. Alzheimer's disease: the amyloid cascade hypothesis. Science 1992; 256 (5054): 184-185
  • 86 Collins-Praino LE, Corrigan F. Does neuroinflammation drive the relationship between tau hyperphosphorylation and dementia development following traumatic brain injury?. Brain Behav Immun 2017; 60: 369-382
  • 87 Sheng JG, Zhu SG, Jones RA, Griffin WS, Mrak RE. Interleukin-1 promotes expression and phosphorylation of neurofilament and tau proteins in vivo. Exp Neurol 2000; 163 (02) 388-391
  • 88 Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci 2003; 23 (05) 1605-1611
  • 89 Ghosh S, Wu MD, Shaftel SS. , et al. Sustained interleukin-1β overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer's mouse model. J Neurosci 2013; 33 (11) 5053-5064
  • 90 Shi JQ, Shen W, Chen J. , et al. Anti-TNF-α reduces amyloid plaques and tau phosphorylation and induces CD11c-positive dendritic-like cell in the APP/PS1 transgenic mouse brains. Brain Res 2011; 1368: 239-247
  • 91 Quintanilla RA, Orellana DI, González-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res 2004; 295 (01) 245-257
  • 92 Maphis N, Xu G, Kokiko-Cochran ON. , et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain 2015; 138 (Pt 6): 1738-1755