Next Article in Journal
The Efficacy of Honey Compared to Silver Sulfadiazine for Burn Wound Dressing in Superficial and Partial Thickness Burns—A Systematic Review and Meta-Analysis
Previous Article in Journal
HIV Prevalence among Injury Patients Compared to Other High-Risk Groups in Tanzania
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Traumatic Brain Injury and Secondary Neurodegenerative Disease

1
Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
2
Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
*
Author to whom correspondence should be addressed.
Trauma Care 2022, 2(4), 510-522; https://doi.org/10.3390/traumacare2040042
Submission received: 8 June 2022 / Revised: 19 September 2022 / Accepted: 20 September 2022 / Published: 26 September 2022

Abstract

:
Traumatic brain injury (TBI) is a devastating event with severe long-term complications. TBI and its sequelae are one of the leading causes of death and disability in those under 50 years old. The full extent of secondary brain injury is still being intensely investigated; however, it is now clear that neurotrauma can incite chronic neurodegenerative processes. Chronic traumatic encephalopathy, Parkinson’s disease, and many other neurodegenerative syndromes have all been associated with a history of traumatic brain injury. The complex nature of these pathologies can make clinical assessment, diagnosis, and treatment challenging. The goal of this review is to provide a concise appraisal of the literature with focus on emerging strategies to improve clinical outcomes. First, we review the pathways involved in the pathogenesis of neurotrauma-related neurodegeneration and discuss the clinical implications of this rapidly evolving field. Next, because clinical evaluation and neuroimaging are essential to the diagnosis and management of neurodegenerative diseases, we analyze the clinical investigations that are transforming these areas of research. Finally, we briefly review some of the preclinical therapies that have shown the most promise in improving outcomes after neurotrauma.

1. Introduction

Neurodegeneration following traumatic brain injury is a complex process that is initiated by several distinct pathways which overwhelm homeostatic stress responses and trigger cellular degeneration and death. Recent studies have demonstrated a progression of neurodegenerative processes months and even years after traumatic brain injury, termed secondary neurodegeneration. Secondary neurodegeneration can manifest in many ways depending on specific etiology and affected neuroanatomy. Chronic traumatic encephalopathy (CTE) is a well-known disease closely associated with repeated traumatic brain injuries (TBI) [1,2]. Parkinson’s disease (PD), frontotemporal dementia (FTD), and other neurodegenerative diseases are less common but can also be induced as a consequence of TBI [3,4,5]. The precise incidence of CTE is hard to quantify due to diagnostic limitations; however, it has gained notoriety due to the prominence of repeated mild TBI in professional sports [1,6]. On the other hand, severe TBI with greater than 1 h loss of consciousness triples the risk of eventually developing PD [3]. Investigation into neurodegenerative disease secondary to TBI is rapidly evolving due to its complex pathophysiology and important public health implications. This review will briefly summarize the current body of knowledge on secondary neurodegeneration, review important imaging modalities related to its diagnosis and management, discuss how the behavioral manifestations of secondary neurodegeneration can aid in diagnosis, and introduce emerging therapeutic targets for the treatment of these diseases.

2. Mechanisms of Neurodegeneration after TBI

The inciting mechanisms of secondary neurodegeneration after TBI are an interdependent set of pathological changes initiated by the primary traumatic injury (Figure 1). The temporal evolution of brain injury after TBI is multidimensional and complex but can be conceptualized as overlapping phases. The “acute injury” phase after TBI is characterized by the predominance of mechanical damage resulting from the initial trauma while the “secondary injury” phase is characterized by the delayed emergence of dysregulated metabolism and inflammation pathways [7,8,9]. The acute phase is generally defined as the first week post-TBI before transitioning into the secondary injury phase that can last months to years [7,10]. Some also advocate for a “subacute” phase as an intermediary that occurs up to 3 months post-TBI [11]. In any case, oxidative stress seems to be a key mediator in the secondary injury phase, as glutamate excitotoxicity, mitochondria dysfunction, and endoplasmic reticulum (ER) stress all contribute to increased reactive oxygen species (ROS) [12,13,14]. Depolarization of glutaminergic neurons after TBI results in increased calcium ion influx through NMDA and AMPA receptors [15]. Excess intracellular calcium increases mitochondrial ROS production through several mechanisms including activation of Ca2+-calmodulin pathways and disruption of the electron transport chain [16]. Endogenous oxidative stress responses are coordinated by the transcription factor Nrf2 [17]. Nrf2 promotes the expression of many cytoprotective proteins including HO-1, NQO-1, and GCLM, among others. These systems can be overwhelmed and become insufficient to prevent ROS-mediated cellular injury [18]. ROS can cause protein damage and misfolding (discussed further below) but may also be especially harmful through lipid peroxidation [19]. Dysregulation of membrane structures such as caveolae in mice is associated with increased markers of neurodegeneration and neuroinflammation [20]. Increased tissue markers of oxidative stress including lipid peroxidation have been observed as far as 12 weeks post-TBI in rats, indicating these pathological mechanisms do not resolve in the acute phase after TBI [21].
Another central element of secondary neurodegeneration is pathological changes to neuronal cytoskeletal dynamics. Axons, especially within the white matter, are particularly susceptible to damage from tensile strain during traumatic injuries due to their unique cellular anatomy [22]. The cytoskeleton of these axons can be completely severed during trauma; however, axonal transport may be disrupted even with mild cytoskeletal damage [22]. This type of injury, often termed diffuse axonal injury (DAI) is common after TBI; however, is likely underreported due to the limitations of imaging techniques and the inability to perform brain biopsies in this patient population [23]. Disrupted axonal transport is one of several mechanisms that impede neuronal homeostatic mechanisms and lead to activation of neuroinflammatory pathways (NFκB– and inflammasome-mediated increases in IL-1β, IL-6, TNFα, etc.) and cell death (caspase-3-mediated apoptosis) [24,25,26]. Pro-inflammatory signals begin locally in damaged neurons but quickly promote reactive gliosis and widespread propagation of the neuroinflammatory cascades by microglia and astrocytes [10,24]. Vascular tissues affected by ROS and pro-inflammatory cytokines are at risk of defective autoregulatory function which can decrease cerebral blood flow and compound cerebral injury [10,25]. These pathological changes result in high protein turnover, particularly in neurons, and may alter ER function by stressing proteostatic mechanisms [27]. ER stress, particularly activation of the unfolded protein response (UPR), is a critical mediator of neurodegenerative change [12,28]. Proteinopathies that occur as a result of misfolding including tauopathies, amyloid plaques, Lewy bodies, and TDP-43 have all been observed after TBI [29,30,31]. Severe (i.e., associated with >1 h loss of consciousness) TBI triples the risk of developing of Lewy bodies in the substantia nigra [3]. ROS can be both a product and contributing factor to axonal degeneration and neuroinflammation, highlighting the interconnections between mechanisms of secondary neurodegeneration.

3. Imaging

Neuroimaging can be used to identify chronic pathological changes from TBI in addition to the acute injury. Generally, TBI disrupts white matter connections and results in cerebral atrophy [32]. This finding tends to be worst in frontotemporal and limbic areas [33], possibly due to trends in traumatic injury mechanisms [34]. Serial quantitative T1 magnetic resonance imaging (MRI) can evaluate neurodegeneration after TBI in a sensitive but non-specific way by assessing cerebral atrophy and volume loss. An increasing ventricle-to-brain ratio is associated with chronic cerebral atrophy in those with TBI after resolution of the acute phase (weeks to months) [35,36]. Generally, TBI-induced neurodegeneration leads to cerebral volumes comparable to that of older individuals with other neurodegenerative diseases; both demonstrating a yearly loss of 1.5% of cerebral volume occurring mostly in sulci and white matter tracts [32,37]. The frontotemporal and limbic areas, which are seated on the sharp sphenoid ridge and edge of the tentorium cerebelli, demonstrate the most severe degenerative changes as their location makes them vulnerable to mechanical deformation. Hippocampal atrophy is especially evident within the limbic system considering its location in the medial temporal lobe and high metabolic demand [38,39]. Patients with DAI-type TBI experience white matter degeneration for months to years following the acute injury as evidenced by studies utilizing MRI diffusor tensor imaging (DTI). DTI is a method for detecting structural changes by analyzing the fractional anisotropy (FA), mean diffusivity (MD), and radial diffusivity (RD) of water molecules. TBI with predominant axonal/white matter injury demonstrates reduced FA and increased MD and RD [40,41]. These findings point to demyelination, loss of axonal integrity, and reduced axonal packing and coherence in frontotemporal and limbic structures such as the anterior limb of the internal capsule, corona radiata, optic radiations, and cingulum [42,43]. Furthermore, changes in these DTI indices are associated with poor neuropsychological performance, including executive function, memory, and functional outcomes [44,45,46,47].
Molecular imaging with positron emission tomography is an evolving modality to identify post-traumatic neurodegeneration in vivo in a specific manner. Following TBI, the PET tracer 11C-Pittsburgh compound-B (11C-PiB) binds amyloid-beta (Aβ) in cortical areas, the striatum, and posterior cingulate cortex similar to Alzheimer’s disease (AD). Unlike AD, there are increased Aβ depositions in the cerebellum in TBI [48,49,50]. Additionally, several PET tracers specific to hyperphosphorylated tau (p-tau) previously used in AD are under investigation for use in TBI. FDDNP is the most well-studied biomarker. FDDNP levels are increased in the midbrain, thalamus, pons, and cingulate gyrus and demonstrate lower binding in temporal and parietal regions in military personnel with mild TBI exposure and football players with suspected chronic traumatic encephalopathy compared to patients with AD [51]; however, FDDNP is non-specific as it also binds Aβ. Other PET imaging biomarkers that bind tau include T801, AV1451, and flortaucipir. Studies are generally limited as they have small sample sizes, lack control groups, or are restricted to one subtype of TBI. While cortical tau tracer uptake varies within individuals with CTE-type TBI, studies have demonstrated consistent uptake in the temporal lobe and limbic system [47,52,53,54]. The current use of PET imaging biomarkers remains in the early stages. The Enhancing Neuroimaging Genetics through Meta-Analysis (ENIGMA) consortium is evaluating the efficacy of these imaging modalities alone and in combination with fluid biomarkers, radiogenomics, or with EEG. Furthermore, there is evolving research investigating magnetic resonance spectroscopy (MRS), functional MRI (fMRI), transcranial Doppler (TCD), single photon emission computed tomography (SPECT), and functional near-infrared spectroscopy) [53,55,56]. Early human and rodent studies evaluating the newly discovered glymphatic pathway reveal that mild, repetitive TBI alters glymphatic clearance rates examined with MRI [57,58,59,60].

4. Cognitive and Behavioral Manifestations

Neurotrauma initiates a process of molecular, cellular, and biochemical changes, which subsequently contribute to neuronal damage and death over time. These secondary processes induce damage through apoptosis, inflammation edema, oxidative stress, and mitochondrial dysfunction, which lead to greater damage than the initial insult [61,62]. These downstream effects precipitate long-term consequences, including an increased risk for developing neurodegenerative disorders such as (CTE), Alzheimer disease (AD), unclassified dementia, and Parkinson’s disease later in life [63,64].
The clinical management of neurotrauma patients varies widely based on country, hospital structure, comorbidities, and severity, with over 378 practice algorithms identified in a recent review [65]. Appropriate multimodality teams for management of patients with neurotrauma include nutrition, primary care, psychiatry, neurology, neurosurgery, physiotherapy, occupational therapy, speech and language therapy, social services, and neuropsychology [66]. A neuropsychological evaluation consists of a clinical interview to thoroughly evaluate attention, executive function, processing speed, and memory, to capture any current cognitive deficits. The integration of neuropsychology in early treatment can be ascribed to the early recognition of cognitive and behavioral deficits (Table 1), which have been the most devastating chronic problems faced by neurotrauma patients [67,68]. Early treatment of neurodegenerative diseases is crucial for therapeutic success, so prompt detection of these conditions is essential to improve quality of life [69].
In chronic traumatic encephalopathy after mild, repetitive TBI, cognitive findings may precede, follow, or co-occur with behavioral findings. Cognitive symptoms include impaired concentration and attention, disorientation, confusion, and speech abnormalities [70,71,72]. Behavioral disturbances are often the earliest finding of CTE and can include paranoia, mood swings, apathy, impulsivity, depression, disinhibition, and suicidality [72,73,74]. An early diagnosis of CTE must involve two or more of the following: pyramidal tract disease, extrapyramidal disease, cognitive and/or behavioral impairment. Neuroimaging studies like PET can aid in the diagnosis [75]. Early signs of unclassified dementia after traumatic brain injury also include apathy, agitation, and disinhibition, but these patients also displayed elevated anxiety, on average, 1.9 years before dementia diagnosis [76].
Parkinsonism is a constellation of symptoms that are characteristically observed in PD. Besides presenting with motor symptoms, PD patients also present with dementia, hallucinations, and cognitive decline. There is considerable evidence suggesting that multiple subtypes of TBI accelerates the neuropathology of PD, therefore early signs of PD should be assessed for early intervention [77]. Behavioral changes are observed early in PD, including motivational decline and slowed thinking [78]. While there is conflicting evidence on epidemiological studies regarding TBI-induced Alzheimer’s disease [3], animal models and clinical studies have found a strong link between the two [79]. Depression, cognitive impairment, and memory loss were the first symptoms to appear preceding AD diagnosis [80].

5. Therapeutic Targets

Histological analysis and brain imaging studies have revealed many associations between TBI and neurodegeneration. On the other hand, studies on potential therapies to target these pathologies are more limited due to the length of time that often occurs between TBI and the clinical manifestations of neurodegenerative disease. This gap in knowledge is being addressed primarily through animal studies of potential therapeutic interventions. Preclinical therapeutic targets currently being investigated are summarized in Table 2.

5.1. Oxidative Stress

Antioxidants and other therapeutics which target ROS generation are appealing translational candidates due to the central role of ROS-mediated cellular damage in the pathogenesis of secondary neurodegeneration. One of the early targeted antioxidant therapies was PEGylated superoxide dismutase (PEG-SOD). The premise of this therapy was that supraphysiological concentrations of SOD would allow rapid detoxification of the ROS generated after TBI. A phase II clinical trial found treatment with PEG-SOD reduced poor outcomes (Glasgow Outcome Scale) at 6 months post-TBI [81]. Many other investigations have turned to naturally-occurring antioxidants. Polyphenols, especially flavonoid-type polyphenols, have shown some promise in reducing oxidative stress after neurotrauma. Several different variations of the flavonoid class compound and some non-flavonoid polyphenols such as resveratrol have been reported in animal studies to reduce tissue oxidation in part through activating the Nrf2 pathway [82,83,84,85]. One of the drawbacks of these types of antioxidants is the unclear mechanism of action and bevy of off-target effects. Mitoquinone, a ubiquinone-based molecule modified to preferentially traffic to the inner mitochondrial membrane, solves some of these issues by specifically targeting an organelle known to be dysfunctional after TBI [86]. Mitoquinone treatment in a mouse model of TBI decreased neuronal apoptosis and helped accelerate the antioxidant response of the Nrf2 pathway [87]. Like all preclinical studies, these investigations suffer some limitations. Rodents do not seem to develop neurotrauma-induced neurodegenerative diseases in the same way humans do and studying the long-term consequences of TBI are difficult in short-lived animals.

5.2. Cell-Based Therapies

As traumatic brain injury can lead to permanent neurodegeneration and neuronal cell death, there has been significant interest in cell-based therapies to restore neurological function. Cell-based therapies include the use of different types of stem cells including neural stem cells and mesenchymal stem cells [88,89]. Pre-clinical models investigating the utility of neural stem cells have demonstrated improved functional outcomes including improved motor recovery and reduced cognitive deficits [90,91,92]. In a rodent model of single, severe TBI, treatment with mesenchymal stem cells reduced proinflammatory mediators and increased ant-inflammatory cytokines [93]. Additional studies where mesenchymal stem cells were genetically engineered to overexpress interleukin-10 found enhanced functional recovery after TBI, possibly via alteration of microglial polarization [94].

5.3. Aggregation-Prone Proteins

Traumatic brain injuries have been linked to progressive neurodegenerative proteinopathies such as Parkinson’s disease and Alzheimer’s disease. Thus, pre-clinical models have been used to study the role of aggregation-prone proteins such as Aß and tau proteins and have found that preventing accumulation of these protein pathologies can improve neurocognitive outcomes after mild, repetitive TBI [95,96,97,98,99]. Tau is necessary for normal microtubules function; however, over phosphorylation of tau is associated with proteinopathy development after TBI [100]. Therapies which target phospho-tau protein for degradation are currently being tested with some promising early results [101,102,103]. Similarly, immunization against pathogenic Aß is an emerging strategy to prevent Aß plaque accumulation [97]. Aß may also be targeted by preventing cleavage of amyloid precursor protein (APP) by amyloidogenic ß- and γ-secretases in favor of cleavage by non-amyloidogenic α-secretase [104,105,106]. These therapies which enhance clearance or decrease production of Aß could potentially be combined with Aß-binding molecules that disrupt Aß aggregation [107].

5.4. Neuroinflammation

Neuroinflammation after traumatic brain injury has been implicated in the subsequent development of neurodegenerative processes [25]. Pre-clinical models have been used to study multiple agents to attenuate this neuroinflammatory response. One group of such agents include pharmacological therapies that are traditionally used for their antimicrobial properties. Minocycline attenuates microglial activation and may reduce secondary brain injury while improving long-term functional outcomes after traumatic brain injury [108,109,110]. Doxycycline decreases matrix metalloprotease-9 (MMP-9) activity, thus preventing blood-brain-barrier disruption and microvascular hyperpermeability [111]. Hydroxychloroquine and chloroquine have been shown to reduce neuroinflammation by decreasing microglia activation and blood-brain-barrier disruption following cortical impact TBI in animal models [112,113].
Synthetic peroxisome proliferator-activated receptor agonists are another target. In preclinical studies, treatment with PPAR agonists prevented microglial activation and mitochondrial dysfunction after traumatic brain injury and resulted in smaller lesions [114,115,116]. In a murine model of single, severe traumatic brain injury, treatment with a cannabinoid type 2 receptor agonist decreased neurodegenerative changes. Specifically, treatment attenuated neuron degeneration and blood-brain-barrier permeability and improved behavioral outcomes [117].
In recent years, a growing number of studies have demonstrated the mechanistic role of pyroptosis in neuroinflammatory processes and have identified inflammasomes as a potential therapeutic target [118,119]. In a pre-clinical model of TBI, selective inhibition of the inflammasome resulted in reduced cerebral edema and improved neurological outcomes in association with decreased inflammatory mediators such as caspase-1 and IL-1β [120]. Therapeutic targets for various other downstream neuroinflammatory mediators also have been investigated. Interferon-beta inhibition reduced neuroinflammation, lesion volume, and long-term neurological impairments in a murine TBI model [121]. Other studies have demonstrated that targeting microglial/macrophage polarization can similarly attenuate neuroinflammation and secondary neurodegenerative processes after TBI [122,123,124,125].
Table 2. Preclinical Studies of Secondary Neurodegeneration Therapies.
Table 2. Preclinical Studies of Secondary Neurodegeneration Therapies.
Therapeutic TargetOutcomes & Mechanism of ActionReferences
Oxidative Stress
PEG-SODCatalyzes the degradation of superoxide radicals; combined with PEG molecules to increase in vivo half-life[64]
Polyphenols & FlavonoidsWater-soluble antioxidants; directly react with ROS in addition to stimulating the Nrf2-ARE pathway[65,66,67,68]
MitoquinoneActs as a renewable antioxidant to reduce mitochondrial ROS [70]
Cell-based Therapies
Neural stem cellsImproves motor recovery and cognition by replacing neurons lost to neurodegeneration[73,74,75]
Mesenchymal stromal stem cellsReduces proinflammatory mediators and improved functional recovery. IL-10 overexpression alters microglial polarization in favor of anti-inflammatory processes[76,77]
Aggregation-prone Proteins
TauPreventing pathologic accumulation via immunization against phosphorylated tau improves neurocognitive outcomes[84,85,86]
Amyloid-beta proteinAβ may be targeted through enhanced clearance (immunization), decreased production (α-secretase overexpression), or decreased aggregation (Aβ binding molecules).[87,88,89,90]
Neuroinflammation
MinocyclineAttenuates microglial activation and improves functional outcomes[92,93,94]
DoxycyclineDecreases MMP-9 activity and preserves blood-brain-barrier integrity[95]
Hydroxychloroquine/chloroquineAttenuates microglial activation and preserves blood-brain-barrier integrity[96,97]
PPAR agonistsAttenuates microglial activation and mitochondrial dysfunction, decreases TBI lesion sizes[98,99,100]
Cannabinoid 2 receptor agonistPrevents neuronal degeneration and preserves blood-brain-barrier integrity[101]
InflammasomesDecreases pro-inflammatory mediators such as caspase-1, IL-18, & IL-1β. Can also reduce pyroptotic cell death by inhibiting gasdermin D cleavage.[102,103,104]
Interferon-betaAttenuates neuroinflammation, decreases lesion volume, and improves long-term functional outcomes[105]

6. Conclusions and Future Directions

Development of neurodegenerative disease after traumatic brain injury represents a complex challenge for providing care to patients who have suffered TBI. The multifactorial pathways that contribute to neurodegeneration are difficult to elucidate, but new discoveries are accelerating progress towards effective diagnosis and treatment of these diseases. Novel imaging technologies and techniques play an important role in this respect, as imaging is critical to initial assessment and long-term tracking of TBI and neurodegeneration. Similarly, understanding how the pathophysiology of neurodegeneration affects neuropsychological pathways is critical to clinical assessment. These investigations also serve the important role of identifying therapeutic targets such as oxidative stress, Proteinopathies, and neuroinflammation. Translational research in this area is crucial to improving long-term outcomes for patients suffering from TBI.
Future inquiry would be most beneficial in addressing the current gaps in knowledge surround TBI and neurodegenerative disease. First, TBI encompasses an enormous array of injury types. There is currently little dedicated literature exploring specific secondary neurodegenerative effects of TBI subtypes such as mild repetitive TBI beyond its association with CTE. Moreover, many studies do not differentiate between specific mechanisms (e.g., crush injury, acceleration/deceleration, penetrating injury, etc.) or anatomical location of primary focal injury. It is reasonable to suspect these variables could impact the secondary neurodegenerative processes that occur after TBI, so future research should explicitly evaluate them.

Author Contributions

Conceptualization, W.S.D., E.J.P., K.P., J.S.H., D.P., B.L.-W.; methodology, W.S.D. and B.L.-W.; software, W.S.D., E.J.P., B.L.-W. writing—original draft preparation, W.S.D., E.J.P., K.P., J.S.H., D.P.; writing—review and editing, W.S.D., E.J.P., B.L.-W.; visualization, W.S.D., E.J.P.; supervision, W.S.D. and B.L.-W.; project administration, W.S.D. and B.L.-W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CTE—chronic traumatic encephalopathy; TBI—traumatic brain injury; PD—Parkinson’s disease; FTD—frontotemporal dementia; AD—Alzheimer’s disease; ER—endoplasmic reticulum; ROS—reactive oxygen species; NMDA—N-methyl-D-aspartic acid; AMDA—α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; Nrf2—nuclear factor erythroid 2-related factor; HO-1—heme oxygenase 1; NQO-1—NAD(P)H Quinone Dehydrogenase 1; GCLM—glutamate cysteine modulatory gene; DAI—diffuse axonal injury; TDP-43—TAR DNA binding protein 43; MRI—magnetic resonance imaging; DTI—diffusor tensor imaging; FA—fractional anisotropy; MD—mean diffusivity; RD—radial diffusivity; FDDNP—2-(1-6-[(2-[fluorine-18]fluoroethyl)(methyl)amino]-2-naphthyl-ethylidene)malononitrile; TCD—transcranial doppler; SPECT—single photon emission computed tomography; PEG—polyethylene glycol; Aβ—amyloid beta; APP—amyloid precursor protein; MMP-9—matrix metalloprotease 9; PPAR—peroxisome proliferator-activated receptor; IL-1β—interleukin 1 beta.

References

  1. Fesharaki-Zadeh, A. Chronic Traumatic Encephalopathy: A Brief Overview. Front. Neurol. 2019, 10, 713. [Google Scholar] [CrossRef] [PubMed]
  2. Raymont, V.; Thayanandan, T. What do we know about the risks of developing dementia after traumatic brain injury? Minerva Med. 2021, 112, 288–297. [Google Scholar] [CrossRef]
  3. Crane, P.K.; Gibbons, L.E.; Dams-O’Connor, K.; Trittschuh, E.; Leverenz, J.B.; Keene, C.D.; Sonnen, J.; Montine, T.J.; Bennett, D.A.; Leurgans, S.; et al. Association between Traumatic Brain Injury and Late Life Neurodegenerative Conditions and Neuropathological Findings. JAMA Neurol. 2016, 73, 1062–1069. [Google Scholar] [CrossRef]
  4. Delic, V.; Beck, K.D.; Pang, K.C.H.; Citron, B.A. Biological links between traumatic brain injury and Parkinson’s disease. Acta Neuropathol. Commun. 2020, 8, 45. [Google Scholar] [CrossRef]
  5. Ramos-Cejudo, J.; Wisniewski, T.; Marmar, C.; Zetterberg, H.; Blennow, K.; de Leon, M.J.; Fossati, S. Traumatic Brain Injury and Alzheimer’s Disease: The Cerebrovascular Link. eBioMedicine 2018, 28, 21–30. [Google Scholar] [CrossRef]
  6. VanItallie, T.B. Traumatic brain injury (TBI) in collision sports: Possible mechanisms of transformation into chronic traumatic encephalopathy (CTE). Metabolism 2019, 100, 153943. [Google Scholar] [CrossRef]
  7. Werner, C.; Engelhard, K. Pathophysiology of traumatic brain injury. Br. J. Anaesth. 2007, 99, 4–9. [Google Scholar] [CrossRef]
  8. Gaetz, M. The neurophysiology of brain injury. Clin. Neurophysiol. 2004, 115, 4–18. [Google Scholar] [CrossRef]
  9. Johnstone, V.P.; Shultz, S.R.; Yan, E.B.; O’Brien, T.J.; Rajan, R. The acute phase of mild traumatic brain injury is characterized by a distance-dependent neuronal hypoactivity. J. Neurotrauma. 2014, 31, 1881–1895. [Google Scholar] [CrossRef]
  10. Risbrough, V.B.; Vaughn, M.N.; Friend, S.F. Role of Inflammation in Traumatic Brain Injury–Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go from Here. Biol. Psychiatry 2022, 91, 438–448. [Google Scholar] [CrossRef]
  11. Mayer, A.R.; Quinn, D.K.; Master, C.L. The spectrum of mild traumatic brain injury: A review. Neurology 2017, 89, 623–632. [Google Scholar] [CrossRef] [PubMed]
  12. Hetz, C.; Saxena, S. ER stress and the unfolded protein response in neurodegeneration. Nat. Rev. Neurol. 2017, 13, 477–491. [Google Scholar] [CrossRef]
  13. Freire, M.A.M. Pathophysiology of neurodegeneration following traumatic brain injury. West Indian Med. J. 2012, 61, 751–755. [Google Scholar]
  14. Cruz-Haces, M.; Tang, J.; Acosta, G.; Fernandez, J.; Shi, R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl. Neurodegener. 2017, 6, 20. [Google Scholar] [CrossRef]
  15. Khatri, N.; Thakur, M.; Pareek, V.; Kumar, S.; Sharma, S.; Datusalia, A.K. Oxidative Stress: Major Threat in Traumatic Brain Injury. CNS Neurol. Disord. Drug Targets 2018, 17, 689–695. [Google Scholar] [CrossRef] [PubMed]
  16. Peng, T.-I.; Jou, M.-J. Oxidative stress caused by mitochondrial calcium overload. Ann. N. Y. Acad. Sci. 2010, 1201, 183–188. [Google Scholar] [CrossRef]
  17. Ma, Q. Role of Nrf2 in Oxidative Stress and Toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef]
  18. Zhang, L.; Wang, H. Targeting the NF-E2-Related Factor 2 Pathway: A Novel Strategy for Traumatic Brain Injury. Mol. Neurobiol. 2018, 55, 1773–1785. [Google Scholar] [CrossRef]
  19. Egawa, J.; Pearn, M.L.; Lemkuil, B.P.; Patel, P.M.; Head, B.P. Membrane lipid rafts and neurobiology: Age-related changes in membrane lipids and loss of neuronal function. J. Physiol. 2016, 594, 4565–4579. [Google Scholar] [CrossRef] [PubMed]
  20. Head, B.P.; Peart, J.N.; Panneerselvam, M.; Yokoyama, T.; Pearn, M.L.; Niesman, I.R.; Bonds, J.A.; Schilling, J.M.; Miyanohara, A.; Headrick, J.; et al. Loss of Caveolin-1 Accelerates Neurodegeneration and Aging. PLoS ONE 2010, 5, e15697. [Google Scholar] [CrossRef]
  21. Webster, K.M.; Wright, D.; Sun, M.; Semple, B.D.; Ozturk, E.; Stein, D.G.; O’Brien, T.; Shultz, S.R. Progesterone treatment reduces neuroinflammation, oxidative stress and brain damage and improves long-term outcomes in a rat model of repeated mild traumatic brain injury. J. Neuroinflamm. 2015, 12, 238. [Google Scholar] [CrossRef] [Green Version]
  22. Johnson, V.E.; Stewart, W.; Smith, D.H. Axonal pathology in traumatic brain injury. Exp. Neurol. 2013, 246, 35–43. [Google Scholar] [CrossRef]
  23. Jang, S.H. Diagnostic Problems in Diffuse Axonal Injury. Diagnostics 2020, 10, 117. [Google Scholar] [CrossRef]
  24. Lozano, D.; Schimmel, S.J.; Acosta, S. Neuroinflammation in traumatic brain injury: A chronic response to an acute injury. Brain Circ. 2017, 3, 135–142. [Google Scholar] [CrossRef] [PubMed]
  25. Pearn, M.L.; Niesman, I.R.; Egawa, J.; Sawada, A.; Almenar-Queralt, A.; Shah, S.B.; Duckworth, J.L.; Head, B.P. Pathophysiology Associated with Traumatic Brain Injury: Current Treatments and Potential Novel Therapeutics. Cell. Mol. Neurobiol. 2017, 37, 571–585. [Google Scholar] [CrossRef]
  26. O’Brien, W.T.; Pham, L.; Symons, G.F.; Monif, M.; Shultz, S.R.; McDonald, S.J. The NLRP3 inflammasome in traumatic brain injury: Potential as a biomarker and therapeutic target. J. Neuroinflamm. 2020, 17, 104–112. [Google Scholar] [CrossRef]
  27. Gerakis, Y.; Hetz, C. Emerging roles of ER stress in the etiology and pathogenesis of Alzheimer’s disease. FEBS J. 2018, 285, 995–1011. [Google Scholar] [CrossRef] [PubMed]
  28. Shah, S.Z.A.; Zhao, D.; Khan, S.H.; Yang, L. Unfolded Protein Response Pathways in Neurodegenerative Diseases. J. Mol. Neurosci. 2015, 57, 529–537. [Google Scholar] [CrossRef]
  29. Washington, P.M.; Villapol, S.; Burns, M.P. Polypathology and Dementia After Brain Trauma: Does Brain Injury Trigger Distinct Neurodegenerative Diseases, or Should They Be Classified Together as Traumatic Encephalopathy? Exp. Neurol. 2016, 275 Pt 3, 381–388. [Google Scholar] [CrossRef]
  30. Uryu, K.; Chen, X.-H.; Martinez, D.; Browne, K.D.; Johnson, V.E.; Graham, D.I.; Lee, V.M.-Y.; Trojanowski, J.Q.; Smith, D.H. Multiple proteins implicated in neurodegenerative diseases accumulate in axons after brain trauma in humans. Exp. Neurol. 2007, 208, 185–192. [Google Scholar] [CrossRef]
  31. Johnson, V.E.; Stewart, W.; Smith, D.H. Widespread τ and amyloid-β pathology many years after a single traumatic brain injury in humans. Brain Pathol. 2012, 22, 142–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Maxwell, W.L.; MacKinnon, M.-A.; Stewart, J.E.; Graham, D.I. Stereology of cerebral cortex after traumatic brain injury matched to the Glasgow Outcome Score. Brain 2010, 133, 139–160. [Google Scholar] [CrossRef] [PubMed]
  33. Adams, J.H.; Doyle, D.; Ford, I.; Gennarelli, T.A.; Graham, D.I.; McLellan, D.R. Diffuse axonal injury in head injury: Definition, diagnosis and grading. Histopathology 1989, 15, 49–59. [Google Scholar] [CrossRef]
  34. Hofman, P.A.; Verhey, F.R.; Wilmink, J.T.; Rozendaal, N.; Jolles, J. Brain lesions in patients visiting a memory clinic with postconcussional sequelae after mild to moderate brain injury. J. Neuropsychiatry Clin. Neurosci. 2002, 14, 176–184. [Google Scholar] [CrossRef]
  35. Bigler, E.D.; Tate, D.F. Brain volume, intracranial volume, and dementia. Investig. Radiol. 2001, 36, 539–546. [Google Scholar] [CrossRef]
  36. Mamere, A.E.; Saraiva, L.A.L.; Matos, A.L.M.; Carneiro, A.A.O.; Santos, A.C. Evaluation of Delayed Neuronal and Axonal Damage Secondary to Moderate and Severe Traumatic Brain Injury Using Quantitative MR Imaging Techniques. Am. J. Neuroradiol. 2009, 30, 947–952. [Google Scholar] [CrossRef] [PubMed]
  37. Graham, N.S.; Sharp, D.J. Understanding neurodegeneration after traumatic brain injury: From mechanisms to clinical trials in dementia. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1221–1233. [Google Scholar] [CrossRef]
  38. Bigler, E.D. Anterior and middle cranial fossa in traumatic brain injury: Relevant neuroanatomy and neuropathology in the study of neuropsychological outcome. Neuropsychology 2007, 21, 515–531. [Google Scholar] [CrossRef]
  39. De La Plata, C.D.M.; Garces, J.; Kojori, E.S.; Grinnan, J.; Krishnan, K.; Pidikiti, R.; Spence, J.; Devous, M.D.; Moore, C.; McColl, R.; et al. Deficits in Functional Connectivity of Hippocampal and Frontal Lobe Circuits After Traumatic Axonal Injury. Arch. Neurol. 2011, 68, 74–84. [Google Scholar]
  40. Veeramuthu, V.; Narayanan, V.; Kuo, T.L.; Delano-Wood, L.; Chinna, K.; Bondi, M.W.; Waran, V.; Ganesan, D.; Ramli, N. Diffusion Tensor Imaging Parameters in Mild Traumatic Brain Injury and Its Correlation with Early Neuropsychological Impairment: A Longitudinal Study. J. Neurotrauma 2015, 32, 1497–1509. [Google Scholar] [CrossRef]
  41. Farbota, K.D.; Bendlin, B.B.; Alexander, A.L.; Rowley, H.A.; Dempsey, R.J.; Johnson, S.C. Longitudinal diffusion tensor imaging and neuropsychological correlates in traumatic brain injury patients. Front. Hum. Neurosci. 2012, 6, 160. [Google Scholar] [CrossRef] [PubMed]
  42. Hellyer, P.J.; Leech, R.; Ham, T.E.; Bonnelle, V.; Sharp, D.J. Individual prediction of white matter injury following traumatic brain injury. Ann. Neurol. 2013, 73, 489–499. [Google Scholar] [CrossRef] [PubMed]
  43. Holleran, L.; Kim, J.H.; Gangolli, M.; Stein, T.; Alvarez, V.; McKee, A.; Brody, D.L. Axonal disruption in white matter underlying cortical sulcus tau pathology in chronic traumatic encephalopathy. Acta Neuropathol. 2017, 133, 367–380. [Google Scholar] [CrossRef]
  44. Palacios, E.M.; Sala-Llonch, R.; Junque, C.; Roig, T.; Tormos, J.M.; Bargallo, N.; Vendrell, P. White matter integrity related to functional working memory networks in traumatic brain injury. Neurology 2012, 78, 852–860. [Google Scholar] [CrossRef]
  45. Wang, J.Y.; Bakhadirov, K.; Abdi, H.; Devous, M.D.; De La Plata, C.D.M.; Moore, C.; Madden, C.J.; Diaz-Arrastia, R. Longitudinal changes of structural connectivity in traumatic axonal injury. Neurology 2011, 77, 818–826. [Google Scholar] [CrossRef] [PubMed]
  46. Sidaros, A.; Engberg, A.W.; Sidaros, K.; Liptrot, M.G.; Herning, M.; Petersen, P.; Paulson, O.B.; Jernigan, T.L.; Rostrup, E. Diffusion tensor imaging during recovery from severe traumatic brain injury and relation to clinical outcome: A longitudinal study. Brain 2008, 131, 559–572. [Google Scholar] [CrossRef] [PubMed]
  47. Stern, R.A.; Adler, C.H.; Chen, K.; Navitsky, M.; Luo, J.; Dodick, D.W.; Alosco, M.L.; Tripodis, Y.; Goradia, D.D.; Martin, B.; et al. Tau Positron-Emission Tomography in Former National Football League Players. N. Engl. J. Med. 2019, 380, 1716–1725. [Google Scholar] [CrossRef]
  48. Hong, Y.T.; Veenith, T.; Dewar, D.; Outtrim, J.G.; Mani, V.; Williams, C.; Pimlott, S.; Hutchinson, P.J.; Tavares, A.; Canales, R.; et al. Amyloid imaging with carbon 11-labeled Pittsburgh compound B for traumatic brain injury. JAMA Neurol. 2014, 71, 23–31. [Google Scholar] [CrossRef]
  49. Mielke, M.M.; Savica, R.; Wiste, H.J.; Weigand, S.D.; Vemuri, P.; Knopman, D.S.; Lowe, V.J.; Roberts, R.O.; Machulda, M.M.; Geda, Y.E.; et al. Head trauma and in vivo measures of amyloid and neurodegeneration in a population-based study. Neurology 2014, 82, 70–76. [Google Scholar] [CrossRef]
  50. Scott, G.; Ramlackhansingh, A.F.; Edison, P.; Hellyer, P.; Cole, J.; Veronese, M.; Leech, R.; Greenwood, R.J.; Turkheimer, F.E.; Gentleman, S.M.; et al. Amyloid pathology and axonal injury after brain trauma. Neurology 2016, 86, 821–828. [Google Scholar] [CrossRef]
  51. Chen, S.T.; Siddarth, P.; Merrill, D.A.; Martinez, J.; Emerson, N.D.; Liu, J.; Wong, K.-P.; Satyamurthy, N.; Giza, C.C.; Huang, S.-C.; et al. FDDNP-PET Tau Brain Protein Binding Patterns in Military Personnel with Suspected Chronic Traumatic Encephalopathy1. J. Alzheimer’s Dis. 2018, 65, 79–88. [Google Scholar] [CrossRef] [PubMed]
  52. Lee, B.; Leavitt, M.J.; Bernick, C.B.; Leger, G.C.; Rabinovici, G.; Banks, S.J. A Systematic Review of Positron Emission Tomography of Tau, Amyloid Beta, and Neuroinflammation in Chronic Traumatic Encephalopathy: The Evidence to Date. J. Neurotrauma 2018, 35, 2015–2024. [Google Scholar] [CrossRef]
  53. Pierre, K.; Dyson, K.; Dagra, A.; Williams, E.; Porche, K.; Lucke-Wold, B. Chronic Traumatic Encephalopathy: Update on Current Clinical Diagnosis and Management. Biomedicines 2021, 9, 415. [Google Scholar] [CrossRef] [PubMed]
  54. Barrio, J.R.; Small, G.W.; Wong, K.P.; Huang, S.C.; Liu, J.; Merrill, D.A.; Giza, C.C.; Fitzsimmons, R.P.; Omalu, B.; Bailes, J.; et al. In Vivo characterization of chronic traumatic encephalopathy using [F-18]FDDNP PET brain imaging. Proc. Natl. Acad. Sci. USA 2015, 112, E2039–E2047. [Google Scholar] [CrossRef] [PubMed]
  55. Olsen, A.; Babikian, T.; Bigler, E.D.; Caeyenberghs, K.; Conde, V.; Dams-O’Connor, K.; Dobryakova, E.; Genova, H.; Grafman, J.; Håberg, A.K.; et al. Toward a global and reproducible science for brain imaging in neurotrauma: The ENIGMA adult moderate/severe traumatic brain injury working group. Brain Imaging Behav. 2021, 15, 526–554. [Google Scholar] [CrossRef] [PubMed]
  56. Amyot, F.; Arciniegas, D.B.; Brazaitis, M.P.; Curley, K.C.; Diaz-Arrastia, R.; Gandjbakhche, A.; Herscovitch, P.; Hinds, S.R., 2nd; Manley, G.T.; Pacifico, A.; et al. A Review of the Effectiveness of Neuroimaging Modalities for the Detection of Traumatic Brain Injury. J. Neurotrauma 2015, 32, 1693–1721. [Google Scholar] [CrossRef]
  57. Christensen, J.; Wright, D.K.; Yamakawa, G.R.; Shultz, S.R.; Mychasiuk, R. Repetitive Mild Traumatic Brain Injury Alters Glymphatic Clearance Rates in Limbic Structures of Adolescent Female Rats. Sci. Rep. 2020, 10, 6254. [Google Scholar]
  58. Piantino, J.; Schwartz, D.L.; Luther, M.; Newgard, C.; Silbert, L.; Raskind, M.; Pagulayan, K.; Kleinhans, N.; Iliff, J.; Peskind, E. Link between Mild Traumatic Brain Injury, Poor Sleep, and Magnetic Resonance Imaging: Visible Perivascular Spaces in Veterans. J. Neurotrauma 2021, 38, 2391–2399. [Google Scholar] [CrossRef]
  59. Kaur, J.; Davoodi-Bojd, E.; Fahmy, L.M.; Zhang, L.; Ding, G.; Hu, J.; Zhang, Z.; Chopp, M.; Jiang, Q. Magnetic Resonance Imaging and Modeling of the Glymphatic System. Diagnostics 2020, 10, 344. [Google Scholar] [CrossRef]
  60. Taoka, T.; Naganawa, S. Glymphatic imaging using MRI. J. Magn. Reson. Imaging 2020, 51, 11–24. [Google Scholar] [CrossRef]
  61. Gupta, R.K.; Prasad, S. Age-Dependent Alterations in the Interactions of NF-κB and N-myc with GLT-1/EAAT2 Promoter in the Pericontusional Cortex of Mice Subjected to Traumatic Brain Injury. Mol. Neurobiol. 2016, 53, 3377–3388. [Google Scholar] [CrossRef] [PubMed]
  62. MacFarlane, M.P.; Glenn, T.C. Neurochemical cascade of concussion. Brain Inj. 2015, 29, 139–153. [Google Scholar] [CrossRef] [PubMed]
  63. Esopenko, C.; Levine, B. Aging, Neurodegenerative Disease, and Traumatic Brain Injury: The Role of Neuroimaging. J. Neurotrauma 2015, 32, 209–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Gupta, R.; Sen, N. Traumatic brain injury: A risk factor for neurodegenerative diseases. Rev. Neurosci. 2016, 27, 93–100. [Google Scholar] [CrossRef]
  65. Borgen, I.M.; Røe, C.; Brunborg, C.; Tenovuo, O.; Azouvi, P.; Dawes, H.; Majdan, M.; Ranta, J.; Rusnak, M.; Wiegers, E.J.; et al. Care transitions in the first 6 months following traumatic brain injury: Lessons from the CENTER-TBI study. Ann. Phys. Rehabil. Med. 2021, 64, 101458. [Google Scholar] [CrossRef]
  66. Abdelmalik, P.A.; Draghic, N.; Ling, G.S.F. Management of moderate and severe traumatic brain injury. Transfusion 2019, 59, 1529–1538. [Google Scholar] [CrossRef]
  67. Kemper, B.; Von Wild, K. Neuropsychological fields in early neurotrauma rehabilitation. Zentralbl. Neurochir. 1999, 60, 168–171. [Google Scholar]
  68. Prince, C.; Bruhns, M.E. Evaluation and Treatment of Mild Traumatic Brain Injury: The Role of Neuropsychology. Brain Sci. 2017, 7, 105. [Google Scholar] [CrossRef]
  69. Sheinerman, K.S.; Umansky, S.R. Early detection of neurodegenerative diseases: Circulating brain-enriched microRNA. Cell Cycle. 2013, 12, 1–2. [Google Scholar] [CrossRef]
  70. Tartaglia, M.C.; Hazrati, L.-N.; Davis, K.D.; Green, R.E.A.; Wennberg, R.; Mikulis, D.; Ezerins, L.J.; Keightley, M.; Tator, C. Chronic traumatic encephalopathy and other neurodegenerative proteinopathies. Front. Hum. Neurosci. 2014, 8, 30. [Google Scholar] [CrossRef]
  71. McKee, A.C.; Stein, T.; Nowinski, C.J.; Stern, R.; Daneshvar, D.; Alvarez, V.E.; Lee, H.-S.; Hall, G.; Wojtowicz, S.M.; Baugh, C.; et al. The spectrum of disease in chronic traumatic encephalopathy. Brain 2013, 136, 43–64. [Google Scholar] [CrossRef] [PubMed]
  72. McKee, A.C.; Cantu, R.C.; Nowinski, C.J.; Hedley-Whyte, E.T.; Gavett, B.; Budson, A.E.; Santini, V.E.; Lee, H.-S.; Kubilus, C.A.; Stern, R. Chronic Traumatic Encephalopathy in Athletes: Progressive Tauopathy After Repetitive Head Injury. J. Neuropathol. Exp. Neurol. 2009, 68, 709–735. [Google Scholar] [CrossRef]
  73. Deb, S.; Burns, J. Neuropsychiatric consequences of traumatic brain injury: A comparison between two age groups. Brain Inj. 2007, 21, 301–307. [Google Scholar] [CrossRef] [PubMed]
  74. Rao, V.; Rosenberg, P.; Miles, Q.S.; Patadia, D.; Treiber, K.; Bertrand, M.; Norton, M.; Steinberg, M.; Tschanz, J.; Lyketsos, C. Neuropsychiatric Symptoms in Dementia Patients with and without a History of Traumatic Brain Injury. J. Neuropsychiatry Clin. Neurosci. 2010, 22, 166–172. [Google Scholar] [CrossRef]
  75. Jordan, B.D. The clinical spectrum of sport-related traumatic brain injury. Nat. Rev. Neurol. 2013, 9, 222–230. [Google Scholar] [CrossRef] [PubMed]
  76. Bray, M.J.C.; Richey, L.N.; Bryant, B.R.; Krieg, A.; Jahed, S.; Tobolowsky, W.; LoBue, C.; Peters, M.E. Traumatic brain injury alters neuropsychiatric symptomatology in all-cause dementia. Alzheimer’s Dement. 2021, 17, 686–691. [Google Scholar] [CrossRef] [PubMed]
  77. Padmakumar, S.; Kulkarni, P.; Ferris, C.F.; Bleier, B.S.; Amiji, M.M. Traumatic brain injury and the development of parkinsonism: Understanding pathophysiology, animal models, and therapeutic targets. Biomed. Pharmacother. 2022, 149, 112812. [Google Scholar] [CrossRef]
  78. Anderson, K.E. Behavioral disturbances in Parkinson’s disease. Dialog. Clin. Neurosci. 2004, 6, 323–332. [Google Scholar] [CrossRef]
  79. Srinivasan, G.; Brafman, D.A. The Emergence of Model Systems to Investigate the Link Between Traumatic Brain Injury and Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 813544. [Google Scholar] [CrossRef]
  80. Bature, F.; Guinn, B.; Pang, D.; Pappas, Y. Signs and symptoms preceding the diagnosis of Alzheimer’s disease: A systematic scoping review of literature from 1937 to 2016. BMJ Open 2017, 7, e015746. [Google Scholar] [CrossRef] [PubMed]
  81. Muizelaar, J.P.; Marmarou, A.; Young, H.F.; Choi, S.C.; Wolf, A.; Schneider, R.L.; Kontos, H.A. Improving the outcome of severe head injury with the oxygen radical scavenger polyethylene glycol-conjugated superoxide dismutase: A Phase II trial. J. Neurosurg. 1993, 78, 375–382. [Google Scholar] [CrossRef]
  82. Fang, J.; Wang, H.; Zhou, J.; Dai, W.; Zhu, Y.; Zhou, Y.; Wang, X.; Zhou, M.-L. Baicalin provides neuroprotection in traumatic brain injury mice model through Akt/Nrf2 pathway. Drug Des. Dev. Ther. 2018, 12, 2497–2508. [Google Scholar] [CrossRef]
  83. Shi, Z.; Qiu, W.; Xiao, G.; Cheng, J.; Zhang, N. Resveratrol Attenuates Cognitive Deficits of Traumatic Brain Injury by Activating p38 Signaling in the Brain. Med Sci. Monit. 2018, 24, 1097–1103. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Y.; Zhang, C.; Peng, W.; Xia, Z.; Gan, P.; Huang, W.; Shi, Y.; Fan, R. Hydroxysafflor yellow A exerts antioxidant effects in a rat model of traumatic brain injury. Mol. Med. Rep. 2016, 14, 3690–3696. [Google Scholar] [CrossRef] [Green Version]
  85. Xu, J.; Wang, H.; Ding, K.; Zhang, L.; Wang, C.; Li, T.; Wei, W.; Lu, X. Luteolin provides neuroprotection in models of traumatic brain injury via the Nrf2–ARE pathway. Free Radic. Biol. Med. 2014, 71, 186–195. [Google Scholar] [CrossRef]
  86. 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. [Google Scholar] [CrossRef]
  87. Zhou, J.; Wang, H.; Shen, R.; Fang, J.; Yang, Y.; Dai, W.; Zhu, Y.; Zhou, M. Mitochondrial-targeted antioxidant MitoQ provides neuroprotection and reduces neuronal apoptosis in experimental traumatic brain injury possibly via the Nrf2-ARE pathway. Am. J. Transl. Res. 2018, 10, 1887–1899. [Google Scholar]
  88. Bonilla, C.; Zurita, M. Cell-Based Therapies for Traumatic Brain Injury: Therapeutic Treatments and Clinical Trials. Biomedicines 2021, 9, 669. [Google Scholar] [CrossRef]
  89. Xiong, Y.; Mahmood, A.; Chopp, M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin. J. Traumatol. 2018, 21, 137–151. [Google Scholar] [CrossRef]
  90. Lee, J.-Y.; Acosta, S.; Tuazon, J.P.; Xu, K.; Nguyen, H.; Lippert, T.; Liska, M.G.; Semechkin, A.; Garitaonandia, I.; Gonzalez, R.; et al. Human parthenogenetic neural stem cell grafts promote multiple regenerative processes in a traumatic brain injury model. Theranostics 2019, 9, 1029–1046. [Google Scholar] [CrossRef]
  91. Xiong, L.-L.; Hu, Y.; Zhang, P.; Zhang, Z.; Li, L.-H.; Gao, G.-D.; Zhou, X.-F.; Wang, T.-H. Neural Stem Cell Transplantation Promotes Functional Recovery from Traumatic Brain Injury via Brain Derived Neurotrophic Factor-Mediated Neuroplasticity. Mol. Neurobiol. 2018, 55, 2696–2711. [Google Scholar] [CrossRef]
  92. Haus, D.L.; López-Velázquez, L.; Gold, E.M.; Cunningham, K.M.; Perez, H.; Anderson, A.J.; Cummings, B.J. Transplantation of human neural stem cells restores cognition in an immunodeficient rodent model of traumatic brain injury. Exp. Neurol. 2016, 281, 1–16. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, R.; Liu, Y.; Yan, K.; Chen, L.; Chen, X.-R.; Li, P.; Chen, F.-F.; Jiang, X.-D. Anti-inflammatory and immunomodulatory mechanisms of mesenchymal stem cell transplantation in experimental traumatic brain injury. J. Neuroinflamm. 2013, 10, 571. [Google Scholar] [CrossRef]
  94. Peruzzaro, S.T.; Andrews, M.M.M.; Al-Gharaibeh, A.; Pupiec, O.; Resk, M.; Story, D.; Maiti, P.; Rossignol, J.; Dunbar, G.L. Transplantation of mesenchymal stem cells genetically engineered to overexpress interleukin-10 promotes alternative inflammatory response in rat model of traumatic brain injury. J. Neuroinflamm. 2019, 16, 2. [Google Scholar] [CrossRef]
  95. Bird, S.M.; Sohrabi, H.R.; Sutton, T.A.; Weinborn, M.; Rainey-Smith, S.R.; Brown, B.; Patterson, L.; Taddei, K.; Gupta, V.; Carruthers, M.; et al. Cerebral amyloid-β accumulation and deposition following traumatic brain injury—A narrative review and meta-analysis of animal studies. Neurosci. Biobehav. Rev. 2016, 64, 215–228. [Google Scholar] [CrossRef]
  96. Ojo, J.O.; Mouzon, B.; Algamal, M.; Leary, P.; Lynch, C.; Abdullah, L.; Evans, J.; Mullan, M.; Bachmeier, C.; Stewart, W.; et al. Chronic Repetitive Mild Traumatic Brain Injury Results in Reduced Cerebral Blood Flow, Axonal Injury, Gliosis, and Increased T-Tau and Tau Oligomers. J. Neuropathol. Exp. Neurol. 2016, 75, 636–655. [Google Scholar] [CrossRef]
  97. Ikonomovic, M.D.; Abrahamson, E.E.; Carlson, S.W.; Graham, S.H.; Dixon, C.E. Novel therapies for combating chronic neuropathological sequelae of TBI. Neuropharmacology 2019, 145, 160–176. [Google Scholar] [CrossRef]
  98. Wu, Y.; Wu, H.; Zeng, J.; Pluimer, B.; Dong, S.; Xie, X.; Guo, X.; Ge, T.; Liang, X.; Feng, S.; et al. Mild traumatic brain injury induces microvascular injury and accelerates Alzheimer-like pathogenesis in mice. Acta Neuropathol. Commun. 2021, 9, 74. [Google Scholar] [CrossRef]
  99. Shin, M.-K.; Vázquez-Rosa, E.; Koh, Y.; Dhar, M.; Chaubey, K.; Cintrón-Pérez, C.J.; Barker, S.; Miller, E.; Franke, K.; Noterman, M.F.; et al. Reducing acetylated tau is neuroprotective in brain injury. Cell 2021, 184, 2715–2732.e23. [Google Scholar] [CrossRef] [PubMed]
  100. Albayram, O.; Kondo, A.; Mannix, R.; Smith, C.; Tsai, C.-Y.; Colin, S.; Herbert, M.K.; Qiu, J.; Monuteaux, M.; Driver, J.; et al. Cis P-tau is induced in clinical and preclinical brain injury and contributes to post-injury sequelae. Nat. Commun. 2017, 8, 1000. [Google Scholar] [CrossRef] [PubMed]
  101. Dai, C.-L.; Hu, W.; Tung, Y.C.; Liu, F.; Gong, C.-X.; Iqbal, K. Tau passive immunization blocks seeding and spread of Alzheimer hyperphosphorylated Tau-induced pathology in 3 × Tg-AD mice. Alzheimer’s Res. Ther. 2018, 10, 13. [Google Scholar] [CrossRef]
  102. Braczynski, A.K.; Schulz, J.B.; Bach, J.-P. Vaccination strategies in tauopathies and synucleinopathies. J. Neurochem. 2017, 143, 467–488. [Google Scholar] [CrossRef] [PubMed]
  103. Lu, K.P.; Kondo, A.; Albayram, O.; Herbert, M.K.; Liu, H.; Zhou, X.Z. 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, 1356–1362. [Google Scholar] [CrossRef]
  104. Mockett, B.G.; Richter, M.; Abraham, W.C.; Müller, U.C. Therapeutic Potential of Secreted Amyloid Precursor Protein APPsα. Front. Mol. Neurosci. 2017, 10, 30. [Google Scholar] [CrossRef]
  105. Corrigan, F.; Vink, R.; Blumbergs, P.C.; Masters, C.L.; Cappai, R.; Heuvel, C.V.D. Evaluation of the effects of treatment with sAPPα on functional and histological outcome following controlled cortical impact injury in mice. Neurosci. Lett. 2012, 515, 50–54. [Google Scholar] [CrossRef]
  106. Corrigan, F.; Vink, R.; Blumbergs, P.C.; Masters, C.L.; Cappai, R.; Heuvel, C.V.D. sAPPα rescues deficits in amyloid precursor protein knockout mice following focal traumatic brain injury. J. Neurochem. 2012, 122, 208–220. [Google Scholar] [CrossRef]
  107. Cohen, A.D.; Ikonomovic, M.D.; Abrahamson, E.E.; Paljug, W.R.; DeKosky, S.T.; Lefterov, I.M.; Koldamova, R.P.; Shao, L.; Debnath, M.L.; Mason, N.S.; et al. Anti-Amyloid Effects of Small Molecule Aβ-Binding Agents in PS1/APP Mice. Lett. Drug Des. Discov. 2009, 6, 437. [Google Scholar] [CrossRef]
  108. Celorrio, M.; Shumilov, K.; Payne, C.; Vadivelu, S.; Friess, S.H. Acute minocycline administration reduces brain injury and improves long-term functional outcomes after delayed hypoxemia following traumatic brain injury. Acta Neuropathol. Commun. 2022, 10, 10. [Google Scholar] [CrossRef]
  109. Whitney, K.; Nikulina, E.; Rahman, S.N.; Alexis, A.; Bergold, P.J. Delayed dosing of minocycline plus N-acetylcysteine reduces neurodegeneration in distal brain regions and restores spatial memory after experimental traumatic brain injury. Exp. Neurol. 2021, 345, 113816. [Google Scholar] [CrossRef]
  110. Bye, N.; Habgood, M.D.; Callaway, J.K.; Malakooti, N.; Potter, A.; Kossmann, T.; Morganti-Kossmann, M.C. Transient neuroprotection by minocycline following traumatic brain injury is associated with attenuated microglial activation but no changes in cell apoptosis or neutrophil infiltration. Exp. Neurol. 2007, 204, 220–233. [Google Scholar] [CrossRef]
  111. Robinson, B.D.; Isbell, C.L.; Melge, A.R.; Lomas, A.M.; Shaji, C.A.; Mohan, C.G.; Huang, J.H.; Tharakan, B. Doxycycline prevents blood–brain barrier dysfunction and microvascular hyperpermeability after traumatic brain injury. Sci. Rep. 2022, 12, 5415. [Google Scholar] [CrossRef] [PubMed]
  112. Hu, J.; Wang, X.; Chen, X.; Fang, Y.; Chen, K.; Peng, W.; Wang, Z.; Guo, K.; Tan, X.; Liang, F.; et al. Hydroxychloroquine attenuates neuroinflammation following traumatic brain injury by regulating the TLR4/NF-κB signaling pathway. J. Neuroinflamm. 2022, 19, 71. [Google Scholar] [CrossRef] [PubMed]
  113. Cui, Y.; Li, R.; Cui, C.-M.; Gao, J.-L.; Sun, L.-Q.; Wang, Y.-C.; Wang, K.-J.; Tian, Y.-X.; Cui, J.-Z. Chloroquine exerts neuroprotection following traumatic brain injury via suppression of inflammation and neuronal autophagic death. Mol. Med. Rep. 2015, 12, 2323–2328. [Google Scholar] [CrossRef]
  114. Yonutas, H.M.; Sullivan, P.G. Targeting PPAR isoforms following CNS injury. Curr. Drug Targets 2013, 14, 733–742. [Google Scholar] [CrossRef]
  115. Strosznajder, A.K.; Wójtowicz, S.; Jeżyna, M.J.; Sun, G.Y.; Strosznajder, J.B. Recent Insights on the Role of PPAR-β/δ in Neuroinflammation and Neurodegeneration, and Its Potential Target for Therapy. Neuromol. Med. 2021, 23, 86–98. [Google Scholar] [CrossRef]
  116. Sauerbeck, A.; Gao, J.; Readnower, R.; Liu, M.; Pauly, J.R.; Bing, G.; Sullivan, P.G. Pioglitazone attenuates mitochondrial dysfunction, cognitive impairment, cortical tissue loss, and inflammation following traumatic brain injury. Exp. Neurol. 2011, 227, 128–135. [Google Scholar] [CrossRef]
  117. Amenta, P.S.; Jallo, J.I.; Tuma, R.F.; Elliott, M.B. A cannabinoid type 2 receptor agonist attenuates blood-brain barrier damage and neurodegeneration in a murine model of traumatic brain injury. J. Neurosci. Res. 2012, 90, 2293–2305. [Google Scholar] [CrossRef] [PubMed]
  118. Yang, B.; Zhong, W.; Gu, Y.; Li, Y. Emerging Mechanisms and Targeted Therapy of Pyroptosis in Central Nervous System Trauma. Front. Cell Dev. Biol. 2022, 10, 832114. [Google Scholar] [CrossRef]
  119. Irrera, N.; Russo, M.; Pallio, G.; Bitto, A.; Mannino, F.; Minutoli, L.; Altavilla, D.; Squadrito, F. The Role of NLRP3 Inflammasome in the Pathogenesis of Traumatic Brain Injury. Int. J. Mol. Sci. 2020, 21, 6204. [Google Scholar] [CrossRef]
  120. Ismael, S.; Nasoohi, S.; Ishrat, T. MCC950, the Selective Inhibitor of Nucleotide Oligomerization Domain-Like Receptor Protein-3 Inflammasome, Protects Mice against Traumatic Brain Injury. J. Neurotrauma 2018, 35, 1294–1303. [Google Scholar] [CrossRef]
  121. Barrett, J.P.; Henry, R.; Shirey, K.A.; Doran, S.J.; Makarevich, O.D.; Ritzel, R.; Meadows, V.A.; Vogel, S.N.; Faden, A.I.; Stoica, B.A.; et al. Interferon-β Plays a Detrimental Role in Experimental Traumatic Brain Injury by Enhancing Neuroinflammation That Drives Chronic Neurodegeneration. J. Neurosci. 2020, 40, 2357–2370. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, H.; Zheng, J.; Xu, S.; Fang, Y.; Wu, Y.; Zeng, J.; Shao, A.; Shi, L.; Lu, J.; Mei, S.; et al. Mer regulates microglial/macrophage M1/M2 polarization and alleviates neuroinflammation following traumatic brain injury. J. Neuroinflamm. 2021, 18, 2. [Google Scholar] [CrossRef] [PubMed]
  123. Nathalie, M.; Polineni, S.P.; Chin, C.N.; Fawcett, D.; Clervius, H.; Maria, Q.S.; Legnay, F.; Rego, L.; Mahavadi, A.K.; Jermakowicz, W.J.; et al. Targeting Microglial Polarization to Improve TBI Outcomes. CNS Neurol. Disord. Drug Targets 2021, 20, 216–227. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, X.; Chen, C.; Fan, S.; Wu, S.; Yang, F.; Fang, Z.; Fu, H.; Li, Y. Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway following experimental traumatic brain injury. J. Neuroinflamm. 2018, 15, 116. [Google Scholar] [CrossRef]
  125. Chio, C.-C.; Lin, M.-T.; Chang, C.-P. Microglial activation as a compelling target for treating acute traumatic brain injury. Curr. Med. Chem. 2015, 22, 759–770. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Summary of mechanisms contributing to secondary neurodegeneration following traumatic brain injury.
Figure 1. Summary of mechanisms contributing to secondary neurodegeneration following traumatic brain injury.
Traumacare 02 00042 g001
Table 1. Early behavioral symptoms in different neurotrauma-induced diseases.
Table 1. Early behavioral symptoms in different neurotrauma-induced diseases.
Neurotrauma-Related DiseaseKey Behavioral FeaturesReferences
Chronic traumatic encephalopathyParanoia, mood swings, apathy, impulsivity, depression, and suicidality[70,71,72]
Unclassified dementiaAnxiety, apathy, and possibly agitation/disinhibition[73,74,75,76]
Parkinson’s diseaseMotivational decline and slowed thinking[3,77,78]
Alzheimer’s diseaseDepression, cognitive impairment, memory loss[79,80]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dodd, W.S.; Panther, E.J.; Pierre, K.; Hernandez, J.S.; Patel, D.; Lucke-Wold, B. Traumatic Brain Injury and Secondary Neurodegenerative Disease. Trauma Care 2022, 2, 510-522. https://doi.org/10.3390/traumacare2040042

AMA Style

Dodd WS, Panther EJ, Pierre K, Hernandez JS, Patel D, Lucke-Wold B. Traumatic Brain Injury and Secondary Neurodegenerative Disease. Trauma Care. 2022; 2(4):510-522. https://doi.org/10.3390/traumacare2040042

Chicago/Turabian Style

Dodd, William S., Eric J. Panther, Kevin Pierre, Jairo S. Hernandez, Devan Patel, and Brandon Lucke-Wold. 2022. "Traumatic Brain Injury and Secondary Neurodegenerative Disease" Trauma Care 2, no. 4: 510-522. https://doi.org/10.3390/traumacare2040042

Article Metrics

Back to TopTop