Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2143 2023-01-30 18:38:41 |
2 format Meta information modification 2143 2023-01-31 04:05:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ruchika, F.;  Shah, S.;  Neupane, D.;  Vijay, R.;  Mehkri, Y.;  Lucke-Wold, B. Molecular Progression of Chronic Traumatic Encephalopathy. Encyclopedia. Available online: https://encyclopedia.pub/entry/40615 (accessed on 02 July 2024).
Ruchika F,  Shah S,  Neupane D,  Vijay R,  Mehkri Y,  Lucke-Wold B. Molecular Progression of Chronic Traumatic Encephalopathy. Encyclopedia. Available at: https://encyclopedia.pub/entry/40615. Accessed July 02, 2024.
Ruchika, Fnu, Siddharth Shah, Durga Neupane, Ruddra Vijay, Yusuf Mehkri, Brandon Lucke-Wold. "Molecular Progression of Chronic Traumatic Encephalopathy" Encyclopedia, https://encyclopedia.pub/entry/40615 (accessed July 02, 2024).
Ruchika, F.,  Shah, S.,  Neupane, D.,  Vijay, R.,  Mehkri, Y., & Lucke-Wold, B. (2023, January 30). Molecular Progression of Chronic Traumatic Encephalopathy. In Encyclopedia. https://encyclopedia.pub/entry/40615
Ruchika, Fnu, et al. "Molecular Progression of Chronic Traumatic Encephalopathy." Encyclopedia. Web. 30 January, 2023.
Molecular Progression of Chronic Traumatic Encephalopathy
Edit

Traumatic brain injury (TBI) is one of the leading causes of death and disability among children and adults in America. In addition, the acute morbidity caused by TBI is implicated in the development of devastating neuropsychiatric and neurodegenerative sequela. TBI is associated with the development of a neurodegenerative condition termed ‘Punch Drunk syndrome’ or ‘dementia pugilistica’, and the more renamed ‘chronic traumatic encephalopathy’. Chronic traumatic encephalopathy (CTE) is a slowly progressive neurodegenerative condition caused by a single or repetitive blow to the head. CTE was first described in boxers and was later found to be associated with other contact sports and military combat. It is defined by a constellation of symptoms consisting of mood disorders, cognitive impairment, and memory loss with or without sensorimotor changes. It is also a Tauopathy characterized by the deposition of hyperphosphorylated Tau protein in the form of neurofibrillary tangles, astrocytoma tangles, and abnormal neurites found in clusters around small vessels, typically at the sulcal depths. Oxidative stress, neuroinflammation, and glutaminergic toxicity caused due to the insult play a role in developing this pathology. Additionally, the changes in the brain due to aging also plays an important role in the development of this condition. 

chronic traumatic encephalopathy Tauopathy molecular basis

2. Neuropathology of Chronic Traumatic Encephalopathy (CTE)

Chronic traumatic encephalopathy is diagnosed most accurately by a neuropathological examination of a brain tissue specimen [1]. The classical finding is an accumulation of hyperphosphorylated Tau protein in neurons and glial cells, mainly involving the perivascular areas and preferential deposition in the cortical sulci depths [2][3][4][5][6]. The specific isomer is cis p-Tau and was found to be produced after traumatic injury to the brain causing cell toxicity, synapse, and circuit dysfunction [6][7].
Studies of concussion and post-concussion syndrome in human subjects found that multifocal traumatic axonal injury is most commonly perivascular [8][9][10] with focal clusters of p-Tau in neurofibrillary tangles, pre-tangles, and neurites [11]. Progression is believed to begin with brain trauma, causing some Tau proteins to become dissociated from microtubules in axons via intracellular calcium influx, glutamate receptor-mediated mechanisms excitotoxicity, and kinase activation mediating hyperphosphorylation of intracellular Tau [12]. In addition, the Tau protein, which dissociates from microtubules, may undergo phosphorylation, misfolding, and aggregation and become proteolytically cleaved by calpains and caspases associated with neurotoxicity [13]. Furthermore, astrocytic p-Tau has been associated with age and not years of exposure to repetitive head impacts (RHI).
The production of cis p-Tau is believed to be initiated by ischemia and in regions that experience mechanical strain forces [14]. Ischemia causing oxidative stress might precede Tau deposition, and the lymphatic system might be a channel for the accumulation of p-Tau in the depths of sulci in a perivascular distribution [14]. The importance of isolated subpial p-Tau astrocytes in the depths of sulci without any neurofibrillary tangles near perivascular areas in the underlying cortex is unknown, but subpial p-Tau astrocytes at the deep cortical sulci is not a phenomenon found in normal aging and has been found in the brains of individuals with a history of chronic repetitive brain trauma [2][11]. The isoform profile of Tau and its phosphorylation state in CTE is like that in Alzheimer’s disease [15], and the neuronal p-Tau pathology shows immunoreactivity to both three repeats (3R) and four repeats (4R) Tau [11][16]. The 4R isoform of Tau is mainly expressed in astrocytes in the subpial region of deep sulci [11][16]. However, the neuronal abnormalities in the hippocampus appear to be primarily 4R Tau in CTE [11].
Grossly, the changes in the brain are not common in the early or mild stages of CTE. Lesions may be present in perivascular spaces in the white matter, mainly in the temporal lobe. Some macroscopic changes include reduction in brain weight, gray and white matter atrophy—typically severe in the frontal and anterior temporal lobes, as well as enlargement of the lateral and third ventricles, cavum septum pellucidum, and septal fenestrations. Other features seen are the atrophy of the thalamus, hypothalamus, and mammillary bodies, the thinning of the isthmus of the septum corpus callosum, and the depigmentation of the locus coeruleus and substantia nigra.
TDP 43 is TAR DNA-binding protein 43 and is also accumulated, causing TDP 43 immunoreactive inclusions [3][4]. It translocates from the nucleus to the cytoplasm, where it can become polyubiquitinated and hyperphosphorylated, resulting in the formation of pathological inclusion bodies [17]. These have been identified in patients with Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and chronic traumatic encephalopathy (CTE) patients [5][18].
Amyloid-β (Aβ) deposition has also been associated with CTE [1][3][5] with significant deposition documented in boxers [19] and American football players [20]. Although Aβ plaques have been identified in older adults, they appear earlier in patients with CTE [2][5].

2. Aging and CTE

The effect of aging on the development of CTE is not completely understood. The most common mechanism is cumulative change over time, with the accumulation of toxic substances, functional decline, and DNA alteration. Mitochondria have a central role in the age-related neurodegeneration and pathogenesis of CTE, and numerous studies have shown that mitochondrial changes occur with aging [21][22]. Mutations in mitochondria and associated oxidative stress contribute to the neurodegenerative process characterized by neuronal cell death, and has been described in patients with AD and PD [23].
Specifically, complex IV and V decline with age, leading to oxidative damage that can disrupt DNA and gene expression [22]. For example, changes are seen in COX gene expression that are associated with a pro-inflammatory state and neurodegeneration [24][25]. Mitochondrial mass also appears to change, as a PCR-based study reported increased content with age [26]. Furthermore, ROS accumulation leads to protein carboxylation, lipid peroxidation, and mtDNA oxidation, which are known to play a role in the development of CTE [22].
The endoplasmic reticulum (ER) is a significant site of calcium storage and protein folding. Alterations in the ER environment cause stress-induced ROS production [27]. Studies have indicated that ER stress events are related to mitochondrial ROS production mechanisms within cells [27]. Ca2+ ions released from the ER augment the production of mitochondrial ROS to induce oxidative phosphorylation at the electron transport chain (ETC). Additionally, Ca2+ ions increase cytochrome c release, which impairs electron transfer, altering mitochondrial membrane potential, and increasing the generation of ROS [27]. ER stress can be provoked by TBI. This can lead to altered ER homeostasis and disrupted folding, leading to unfolded proteins and protein aggregates which are detrimental to cell survival [28]. CTE is characterized by hyperphosphorylated Tau protein and, in some patients, amyloid beta-peptide. Neurons containing NFTs showed an increase in levels of free and protein-bound calcium compared with tangle-free neurons [29][30]. Calcium plays an essential role in apoptosis, neurodegeneration, and CTE.
Microglia play a pivotal role in immune surveillance, plasticity, and development [31][32]. They have a dual function, allowing them to switch from a pro-inflammatory state to a neuroreparative state. In acute TBI, the microglia take part in the inflammatory process and help with the clearing of debris. Repetitive trauma does not allow the microglia to switch to their reparative mode. Hence, immunoexcitotoxicity is believed to contribute to the development of CTE [33]. There is evidence of microglia priming in the aged brain. For example, there is increased expression of inflammatory markers, including major histocompatibility complex II and complement receptor 3 (CD11), in the aging human brain [34][35]. There is also an increase in the inflammatory profile in astrocytes with age [36]. The increased inflammatory markers observed on astrocytes and microglia in the aged brain translate to an exaggerated immune response following trauma. This leads to a maladaptive response characterized by amplified and prolonged cytokine production, anorexia, prolonged social withdrawal, depressive behavior, and cognitive impairment, among other symptoms [37]. Primed microglia in an aged brain produce a more robust response to a peripheral stimulus such as stress and trauma. In addition, a study showed that in a focused laser injury mice model, there was slower migration of microglia towards the site of injury, and the microglia were aggregated at the site of damage for a longer duration than that of adult mice [38]. These functionally impaired microglia are senescent or dystrophic and indicate worse outcomes in brain injury.
Another study showed an age-dependent deficiency in the glutamate transporter on neurons of the excitatory amino acid carrier (EAAC1) [39]. It showed that genetically null mice had reduced glutathione levels and, with aging, developed brain atrophy and behavioral changes [39]. Low glutathione levels are linked to increased ROS, and ROS has been implicated in neuronal inflammation and the development of CTE.

3. Neurodegenerative Diseases and CTE

Many studies have assessed the role of TBI in developing neurodegenerative conditions, and meta-analyses of these studies have shown a significant association between TBI and AD [40][41], PD, ALS [42], and FTD (Frontotemporal dementia) [43]. Pathophysiological changes of CTE have been shown to mimic molecular and cellular changes found in other neurodegenerative diseases such as AD and ALS. A study showed that microglia priming had been described in several neurodegenerative conditions, including AD. CD200 receptors are essential for switching microglia from neurodestructive mode to neuroprotective mode. Recent studies have shown reduced CD200 and CD200 microglial receptors in pathologically affected areas in AD. Macrophages of PD patients also have reduced CD200 [44].
Evidence suggests that activated microglia are a chronic source of multiple neurotoxic factors, such as tumor necrosis factor-α, NO (nitric oxide), interleukin-1β, and reactive oxygen species (ROS), driving progressive neuron damage. Microglia can become chronically activated by either a single stimulus (e.g., lipopolysaccharide or neuron damage) or multiple stimuli exposures, resulting in cumulative neuronal loss with time. This explains the prolonged activation of microglia in neurodegenerative conditions [45]. Studies have shown that pro-inflammatory immune stimulation was insufficient to cause brain pathology but triggered extensive neurodegeneration in patients with pre-existing or coexisting brain pathology in the form of excitotoxicity. Morimoto et al. found that injecting LPS plus ibotenate, an NMDA receptor agonist, led to significant neuronal degeneration and severe tissue collapse. By blocking excitotoxicity, tissue damage was prevented, despite substantial microglial activation [46]. If the ibotenate was delayed by a day after the LPS injection, gross microglial activation occurred along with significant neurodegeneration.
Tau NFT deposition is a characteristic finding identified in the brains of AD patients, as well as frontotemporal dementia (FD), Pick’s disease (PiD), and progressive supranuclear palsy (PSP), among others. These tangles have also been distinctively found in the brains of patients suffering from CTE [11]. A study done by Holf et al. discovered that the distribution of these tangles was similar to the ones found in AD and PD. However, in 2018 McKee et al. compared the brains of 68 men who had suffered a CTE to 18 age and sex-matched brains who had not suffered a CTE, discovering that brains who had undergone trauma had a unique NFT distribution that was different from any other tauopathy [2]. These included perivascular NFTs distributed in the cortex and subpial astrocytes at the sulcal depths.
Similarly, AD is also known to have AB plaque deposits around neurons, and 50% of the cases of CTE have also been found to have these deposits. However, they appear at a later disease stage and in a lesser proportion [2]. Furthermore, their presence is commonly associated with faster clinical deterioration, Lewy body formation, dementia, and parkinsonism [2].
TDP-43 is a nuclear protein that regulates the transcription of genes that bind to the E3 ubiquitin ligase Parkin mRNA to regulate its expression. The wild type of this protein has been linked to several neurogenerative diseases, such as Huntington’s disease (HD) and ALS in the hippocampus of patients suffering from AD and the limbic system in PSP (supranuclear palsy), in addition to Lewy body dementia [47]. A study done by McKee et al. studied the brains of 12 athletes aged between 42 and 85 who had developed CTE, finding that three of them had also developed motor neuron disease (MND), similar to the symptoms presented in patients who have ALS [48]. Two of the patients also developed cognitive impairment, dementia, and behavioral changes. Seven of the nine patients that did not develop MND showed TDP-43 immunoreactivity in a specific area of the brain. The athlete’s brains that developed MND all showed TDP-34 immunoreactivity throughout their brains, including the brainstem and spinal cord [48]. These findings were compared to sporadic ALS cases, in which the only difference was that none presented Tau protein tangles similar to those on CTE brains.
Moreover, it has been proven that oxidative stress plays a central role in the neuronal damage produced post-TBI. Antioxidant mechanisms are interfered with due to trauma, leading to the accumulation of ROS due to NADPH oxidase (Nox2) upregulation, which leads to DNA damage and brain inflammation [49]. In the same way, Nox2 has been proven to induce Aβ plaque formation and accumulation, which predisposes AD development. Although the final pathological manifestations of CTE closely resemble that of sporadic AD, there are some differences, especially the predominance of Tau pathology over amyloid accumulation in affected brain regions. In conclusion, CTE is a modifiable risk factor. Efforts towards developing robust biomarkers and well-designed, prospective epidemiological studies involving contact sports players from an early age to assess the risk of neurodegeneration and develop therapies are essential.

References

  1. McKee, A.C. The Neuropathology of Chronic Traumatic Encephalopathy: The Status of the Literature. Semin. Neurol. 2020, 40, 359–369.
  2. Mckee, A.C.; Abdolmohammadi, B.; Stein, T.D. The neuropathology of chronic traumatic encephalopathy. Handb. Clin. Neurol. 2018, 158, 297–307.
  3. Manley, G.; Gardner, A.J.; Schneider, K.J.; Guskiewicz, K.M.; Bailes, J.; Cantu, R.C.; Castellani, R.J.; Turner, M.; Jordan, B.D.; Randolph, C.; et al. A systematic review of potential long-term effects of sport-related concussion. Br. J. Sport. Med. 2017, 51, 969–977.
  4. Baugh, C.M.; Stamm, J.M.; Riley, D.O.; Gavett, B.E.; Shenton, M.E.; Lin, A.; Nowinski, C.J.; Cantu, R.C.; McKee, A.C.; Stern, R.A. Chronic traumatic encephalopathy: Neurodegeneration following repetitive concussive and subconcussive brain trauma. Brain Imaging Behav. 2012, 6, 244–254.
  5. Smith, D.H.; Johnson, V.E.; Trojanowski, J.Q.; Stewart, W. Chronic traumatic encephalopathy—Confusion and controversies. Nat. Rev. Neurol. 2019, 15, 179–183.
  6. Shively, S.B.; Priemer, D.S.; Stein, M.B.; Perl, D.P. Pathophysiology of Traumatic Brain Injury, Chronic Traumatic Encephalopathy, and Neuropsychiatric Clinical Expression. Psychiatr. Clin. N. Am. 2021, 44, 443–458.
  7. VanItallie, T.B. Traumatic brain injury (TBI) in collision sports: Possible mechanisms of transformation into chronic traumatic encephalopathy (CTE). Metabolism 2019, 100, 153943.
  8. Blumbergs, P.C.; Scott, G.; Manavis, J.; Wainwright, H.; Simpson, D.A.; McLean, A.J. Stalning af amyloid percursor protein to study axonal damage in mild head Injury. Lancet 1994, 344, 1055–1056.
  9. McKee, A.C.; Daneshvar, D.H.; Alvarez, V.E.; Stein, T.D. The neuropathology of sport. Acta Neuropathol. 2014, 127, 29–51.
  10. Oppenheimer, D.R. Microscopic lesions in the brain following head injury. J. Neurol. Neurosurg. Psychiatry 1968, 31, 299–306.
  11. Mckee, A.C.; Stein, T.D.; Nowinski, C.J.; Stern, R.A.; Daneshvar, D.H.; Alvarez, V.E.; Lee, H.-S.; Hall, G.; Wojtowicz, S.M.; Baugh, C.M.; et al. The spectrum of disease in chronic traumatic encephalopathy. Brain 2013, 136, 43–64.
  12. Genis, L.; Chen, Y.; Shohami, E.; Michaelson, D.M. Tau hyperphosphorylation in apolipoprotein E-deficient and control mice after closed head injury. J. Neurosci. Res. 2000, 60, 559–564.
  13. Amadoro, G.; Ciotti, M.T.; Costanzi, M.; Cestari, V.; Calissano, P.; Canu, N. NMDA receptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation. Proc. Natl. Acad. Sci. USA 2006, 103, 2892–2897.
  14. Daneshvar, D.H.; Goldstein, L.E.; Kiernan, P.T.; Stein, T.D.; McKee, A.C. Post-traumatic neurodegeneration and chronic traumatic encephalopathy. Mol. Cell. Neurosci. 2015, 66, 81–90.
  15. Schmidt, M.; Zhukareva, V.; Newell, K.; Lee, V.; Trojanowski, J. Tau isoform profile and phosphorylation state in dementia pugilistica recapitulate Alzheimer’s disease. Acta Neuropathol. 2001, 101, 518–524.
  16. Stein, T.D.; Alvarez, V.E.; McKee, A.C. Chronic traumatic encephalopathy: A spectrum of neuropathological changes following repetitive brain trauma in athletes and military personnel. Alzheimer’s Res. Ther. 2014, 6, 4.
  17. Neumann, M.; Kwong, L.K.; Sampathu, D.M.; Trojanowski, J.Q.; Lee, V.M.-Y. TDP-43 Proteinopathy in Frontotemporal Lobar Degeneration and Amyotrophic Lateral Sclerosis. Arch. Neurol. 2007, 64, 1388.
  18. Johnson, V.E.; Stewart, W.; Trojanowski, J.Q.; Smith, D.H. Acute and chronically increased immunoreactivity to phosphorylation-independent but not pathological TDP-43 after a single traumatic brain injury in humans. Acta Neuropathol. 2011, 122, 715–726.
  19. Roberts, G.W.; Allsop, D.; Bruton, C. The occult aftermath of boxing. J. Neurol. Neurosurg. Psychiatry 1990, 53, 373–378.
  20. Mez, J.; Daneshvar, D.H.; Kiernan, P.T.; Abdolmohammadi, B.; Alvarez, V.E.; Huber, B.R.; Alosco, M.L.; Solomon, T.M.; Nowinski, C.J.; McHale, L.; et al. Clinicopathological Evaluation of Chronic Traumatic Encephalopathy in Players of American Football. JAMA 2017, 318, 360.
  21. Lenaz, G.; Merlo Pich, M.; Genova, M.; Ventura, B.; Bovina, C.; Formiggini, G.; Parenti Castelli, G. Mitochondrial bioenergetics in aging. Mitochondrial Bioenerg. Aging Biochim. Biophys. Acta 2000, 1459, 397–404. Available online: www.elsevier.com/locate/bba (accessed on 10 October 2022).
  22. Navarro, A.; Boveris, A. The mitochondrial energy transduction system and the aging process. Am. J. Physiol. Cell Physiol. 2007, 292, 670–686.
  23. Lin, M.T.; Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 2006, 443, 787–795.
  24. Corral-Debrinski, M.; Horton, T. Mitochondrial DNA deletions in human brain: Regional variability and increase with advanced age. Nat. Genet. 1992, 2, 324–329.
  25. Minghetti, L. Cyclooxygenase-2 (COX-2) in Inflammatory and Degenerative Brain Diseases. J. Neuropathol. Exp. Neurol. 2004, 63, 901–910. Available online: https://academic.oup.com/jnen/article/63/9/901/2916516 (accessed on 10 October 2022).
  26. Barrientos, A.; Casademont, J.; Cardellach, F.; Estivill, X.; Urbano-Marquez, A.; Nunes, V. Reduced steady-state levels of mitochondrial RNA and increased mitochondrial DNA amount in human brain with aging. Mol. Brain Res. 1997, 52, 284–289.
  27. Bhandary, B.; Marahatta, A.; Kim, H.R.; Chae, H.J. An involvement of oxidative stress in endoplasmic reticulum stress and its associated diseases. Int. J. Mol. Sci. 2013, 14, 434–456.
  28. Kaufman, R.J.; Scheuner, D.; Schröder, M.; Shen, X.; Lee, K.; Liu, C.Y.; Arnold, S.M. The unfolded protein response in nutrient sensing and differentiation. Nat. Rev. Mol. Cell Biol. 2002, 3, 411–421.
  29. Verkhratsky, A. Physiology and Pathophysiology of the Calcium Store in the Endoplasmic Reticulum of Neurons. Physiol. Rev. 2005, 85, 201–279.
  30. Mattson, M.P. apoptosis in neurodegenerative disorders. Nat. Rev. Mol. Cell Biol. 2000, 1, 120–130.
  31. Ransohoff, R.M.; Perry, V.H. Microglial physiology: Unique stimuli, specialized responses. Annu. Rev. Immunol. 2009, 27, 119–145.
  32. Hanisch, U.K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387–1394.
  33. Blaylock, R.; Maroon, J. Immunoexcitotoxicity as a central mechanism in chronic traumatic encephalopathy-A unifying hypothesis. Surg. Neurol. Int. 2011, 2, 107.
  34. Streit, W.J.; Sammons, N.W.; Kuhns, A.J.; Sparks, D.L. Dystrophic Microglia in the Aging Human Brain. GLIA 2004, 45, 208–212.
  35. Henry, C.J.; Huang, Y.; Wynne, A.M.; Godbout, J.P. Peripheral lipopolysaccharide (LPS) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro-inflammatory IL-1β and anti-inflammatory IL-10 cytokines. Brain Behav. Immun. 2009, 23, 309–317.
  36. Palmer, A.L.; Ousman, S.S. Astrocytes and Aging. Front. Aging Neurosci. 2018, 10, 337.
  37. Koellhoffer, E.C.; Mccullough, L.D.; Ritzel, R.M. Molecular Sciences Old Maids: Aging and Its Impact on Microglia Function. Int. J. Mol. Sci. 2017, 18, 769.
  38. Damani, M.R.; Zhao, L.; Fontainhas, A.M.; Amaral, J.; Fariss, R.N.; Wong, W.T. Age-related alterations in the dynamic behavior of microglia. Aging Cell 2011, 10, 263–276.
  39. Aoyama, K.; Sang, W.S.; Hamby, A.M.; Liu, J.; Wai, Y.C.; Chen, Y.; Swanson, R.A. Neuronal glutathione deficiency and age-dependent neurodegeneration in the EAAC1 deficient mouse. Nat. Neurosci. 2006, 9, 119–126.
  40. Gu, D.; Ou, S.; Liu, G. Traumatic Brain Injury and Risk of Dementia and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. Syst. Rev. Neuroepidemiol. 2021, 56, 4–16.
  41. Zhang, J.; Zhang, Y.; Zou, J.; Caoid, F. A meta-analysis of cohort studies: Traumatic brain injury and risk of Alzheimer’s Disease. PLoS ONE 2021, 16, e0253206.
  42. Chen, S.; Sayana, P.; Zhang, X.; Le, W. Genetics of amyotrophic lateral sclerosis: An update. Mol. Neurodegener. 2013, 28, 8.
  43. Gardner, R.C.; Yaffe, K. Epidemiology of mild traumatic brain injury and neurodegenerative disease. Mol. Cell. Neurosci. 2015, 66, 75–80.
  44. Rabaneda-Lombarte, N.; Manuel Vidal-Taboada, J.; Valente, T.; Ezquerra, M.; Fernández-Santiago, R.; Martí, M.J.; Compta, Y.; Saura, J.; Solà, C. ARTICLE Altered expression of the immunoregulatory ligand-receptor pair CD200-CD200R1 in the brain of Parkinson’s disease patients. Npj Park. Dis. 2022, 8, 27.
  45. Lull, M.E.; Block, M.L. Microglial Activation and Chronic Neurodegeneration. Neurother. J. Am. Soc. Exp. Neurother. 2010, 7, 345–365.
  46. Morimoto, K.; Murasugi, T.; Oda, T. Acute neuroinflammation exacerbates excitotoxicity in rat hippocampus in vivo. Exp. Neurol. 2002, 177, 95–104.
  47. Hebron, M.L.; Lonskaya, I.; Sharpe, K.; Weerasinghe, P.P.K.; Algarzae, N.K.; Shekoyan, A.R.; Moussa, C.E.H. Parkin ubiquitinates tar-DNA binding protein-43 (TDP-43) and promotes its cytosolic accumulation via interaction with histone deacetylase 6 (HDAC6). J. Biol. Chem. 2013, 288, 4103–4115.
  48. McKee, A.C.; Gavett, B.E.; Stern, R.A.; Nowinski, C.J.; Cantu, R.C.; Kowall, N.W.; Perl, D.P.; Tessa Hedley-Whyte, E.; Price, B.; Sullivan, C.; et al. TDP-43 Proteinopathy and Motor Neuron Disease in Chronic Traumatic Encephalopathy. J. Neuropathol. Exp. Neurol. 2010, 69, 918–929. Available online: www.jneuropath.com (accessed on 7 October 2022).
  49. Ismail, H.; Shakkour, Z.; Tabet, M.; Abdelhady, S.; Kobaisi, A.; Abedi, R.; Nasrallah, L.; Pintus, G.; Al-Dhaheri, Y.; Mondello, S.; et al. Traumatic Brain Injury: Oxidative Stress and Novel Anti-Oxidants Such as Mitoquinone and Edaravone. Antioxidants 2020, 9, 943.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 423
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 31 Jan 2023
1000/1000
Video Production Service