European Journal of Neurodegenerative Diseases 2021; 10(2) July-December: 41-50


INFLAMMATION IN TRAUMATIC BRAIN INJURY: THE RELATIONSHIP BETWEEN MICROGLIA AND NEURODEGENERATIVE DISEASES

M. Trimarchi

Department of Medicine and Surgery, Centre of Neuroscience of Milan, University of Milan, Italy

*Correspondence to: trimarchi.matteo@hsr.it

Received: 28 September, 2021
Accepted: 27 December, 2021
 
2279-5855 (2021)
Copyright © by BIOLIFE
This publication and/or article is for individual use only and may not be further
reproduced without written permission from the copyright holder.
Unauthorized reproduction may result in financial and other penalties
Disclosure: all authors report no conflicts of interest relevant to this article.

ABSTRACT

 

Traumatic brain injury (TBI) is responsible for a high prevalence of global death, disability, and morbidity, and the effects for patients and their families can be devastating, with even a mild, non-concussive injury able to have long-term effects. Microglia, the innate immune cells resident in the central nervous system (CNS), respond swiftly after TBI and are important for reparation, but also secrete pro-inflammatory cytokines, which participate in inflammation. Heightened microglial activation can result in “primed” microglia and chronic neuroinflammation, which can lead to cognitive impairment and the development of progressive neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), or chronic traumatic encephalopathy (CTE). These disorders include the abnormal accumulation of phosphorylated tau proteins and amyloid-β (Aβ) peptide deposits in the brain, with studies finding neuroinflammation and microgliosis to be linked to neuronal damage, disease worsening, and outcome. Microglial manipulation and neuroimaging are being used to define the role of microglia in these neurodegenerative diseases, develop treatments, and distinguish the correct time frame for therapeutic intervention.

 

Keywords: TBI, Inflammation, Neuroinflammation, Neurodegenerative, Disease, Microglia, Immune, AD, PD, CTE

  

INTRODUCTION

 

Traumatic brain injury (TBI) is defined by the Centers for Disease Control and Prevention as “a disruption in the normal function of the brain that can be caused by a bump, blow, or jolt to the head, or penetrating head injury” (1). Prevalent causes of TBIs include falls, motor vehicle accidents, strikes with objects, and assault (2). TBIs can affect anyone, but a higher incidence has been reported in children, adolescents, adults over 75 years of age, and males (3). In addition, individuals who play contact sports and military service members are at higher risk due to repetitive injury over a sustained time.

TBIs are a major source of death and morbidity worldwide and can cause lifelong consequences. According to the Glasgow Coma Scale, injuries can be classified as mild, moderate, or severe, depending on the time of unconsciousness, mental state, and posttraumatic amnesia (4). The worldwide prevalence of TBI is great, considering that 2.8 million people were diagnosed in the United States alone in 2013 (2), but the true numbers are difficult to define as many mild cases go unreported.

TBI can produce neuronal, functional, and inflammatory consequences. The inflammatory response following TBI is a mix of complex, superimposed mechanisms, and age, sex, mechanism and degree of injury, secondary insults, and genetic variation are all factors that can impact that event (5)(6). Even after mild injury, TBI can cause chronic neuroinflammation and lead to neuropsychiatric and neurodegenerative pathologies months and years after the injury, a time of complex neuroinflammatory cascades. In addition, studies have shown that cognitive decline affects between 15 and 30% of TBI patients (7-9), and even subconcussive injury can result in long-term neurologic consequences (10).

The immune system reacts to protect the body against injury and infection, which it performs by engaging in inflammatory challenges in response to noxious stimuli. After TBI, the immune response is essential for the repair process; however, an upregulated immune response can attack healthy cells and cause damaging, chronic neuroinflammation.

The initial injury that occurred in TBI is considered the “primary injury”, and the multifaceted pathophysiological reactions that follow comprise the “secondary injury”, the period when neuroinflammatory processes are activated and may continue to become unresolved microglia-mediated inflammation (11). These primary and secondary TBI injuries can result in neuropathology (12). In addition, microglia-mediated inflammation can continue in this time of secondary injury long after the primary injury was incurred, contributing to chronic neuroinflammation, which has been seen in human TBI patients and animal models.

The role of post-TBI neuroinflammation in the development and progression of neurodegenerative disorders has become an interesting and promising avenue of exploration and can be useful for treatment as recent research continues to shed light on their connection. This review aims to summarize the role of microglia in TBI-induced neuroinflammation, focusing on the role they play in Alzheimer’s Disease (AD), Parkinson’s Disease (PD), and chronic traumatic encephalopathy (CTE), progressive neurodegenerative disorders which are being studied for this theme. AD, PD, and CTE  include the following characterizations: abnormal accumulation of phosphorylated tau proteins and the formation of amyloid-β (Aβ) peptide deposits in the brain.

Microglia activation in TBI

Immune function is greatly influenced by TBI, with an increased inflammatory response, and repetitive events can be magnifying, adding more inflammation into an already activated system (13).

Neuroinflammation occurs in the central nervous system (CNS), which comprises the brain, spinal cord, and optic nerves. Microglia, a glial component of the CNS, derive from the progenitors of mononuclear myeloid cells and respond rapidly after TBI has occurred, migrating to the source of injury within 30 minutes (14). Microglia are important for immune surveillance, mediating the innate immune capacity of the CNS with synaptic pruning, debris clearance, and tissue protection. They have a similar function to macrophages, and when activated, microglia secrete various harmful compounds to the body, including pro-inflammatory cytokines such as interleukin-6 (IL-6), and interleukin 1β (IL-1β), Tumor Necrosis Factor (TNF), and other inflammatory mediators (15). Innate immune inflammatory cytokines act on cytokine receptors and Toll-like receptors.

Microglia are ubiquitous throughout the CNS and are “quiet” in their stable microenvironment. Any detected insult or stimuli can initiate immediate response, leading to “microglial activation”. Microglial “priming” occurs successively when microglia are sensitized and overly responsive to stimuli (16). After TBI, increased sensitization of microglia show raised levels of innate immune markers (17). The disruption to microglial homeostasis that occurs after a TBI can continue for months or years; this has been shown in different studies that showed increased Iba1, CD68+, MHCII, and CD68 labeled microglia after injury (18-21).

Detection of activated microglia bound to positron emission tomography (PET)-detectable ligands can indicate the level of inflammation after TBI, which was seen up to 17 years after injury and was related to impaired cognitive effects (22)(23). In a study of former professional football players who endured repetitive TBI throughout their careers, activated microglia and macrophages were shown after retirement and before the decline in cognitive processing (24). In rodent models, increased microglia labeling and white matter damage affected hippocampal-dependent learning (19).

Serum cytokines have also shown chronic hyper-activation following TBI, with elevated TNF in serum expressed post-injury, which has been linked to poor neuropsychiatric outcomes (25). The mitogen-activated protein kinase (MAPK) p38α responds to stress stimuli, controlling diverse cellular processes with unique functions (26). After diffuse TBI, p38α MAPK signaling in microglia has been seen to promote cytokine production, therefore perpetuating TBI-induced microglial activation; furthermore, microglial depletion in mice was associated with less synaptic protein loss and motor deficits, suggesting that this microglial signaling pathway may be related to the development of neuropathology post-TBI (12).

Chronic neuroinflammation after TBI should be contained if microglia response can be confined to the acute TBI stage and limited during the secondary inflammatory stage, a process that can be neuroprotective (12).

Prolonged disruption of microglial homeostasis induces neuroinflammation in the CNS and can lead to neuronal damage. However, microglial depletion has been shown to prevent detrimental gene suppression, not during the acute phase of TBI but during the dynamic inflammatory stage that follows.

Microglia are plastic, changing in appearance depending on their function (27, 28). Neurons and microglia display dynamic structural associations after TBI in a species-dependent manner (29). In a porcine TBI model, microglia were activated within 15 minutes, and reactivity was focused proximal to individual injured neurons (30). CD68+ microglial cells increased in number from the first day following injury until the 28th (31). TBI also initiates rod microglia morphology and unique phenotype (18)(32)(33). In a study by Ziebell et al., microglia, remodeling by structural transition, were seen to become rod-shaped in the rodent cortex at one-week post-midline fluid percussion injury, retained rod morphology for no less than 4 weeks, and the formation was dependent on the presence of preserved neuronal tissue (18).

Post TBI, microglia-mediated inflammation can cause neuronal dysfunction, affecting plasticity and connectivity (23). However, a study by Witcher et al. showed that microglial depletion prevented detrimental gene suppression post-TBI, preventing TBI-associated cognitive impairments in mice (23).

A history of TBI especially repeated events, is a risk factor for the development and severity of different neurodegenerative diseases (13). Chronic neuroinflammation, and the continuous overly active state of primed microglia that follows TBI, have been implicated in AD, PD, CTE, Huntington’s Disease, vascular dementia, and depression (34)(35). Microglial activation can lead to the production of Aβ, tau pathology, and neuroinflammation, along with a reduced release of neurotrophic factors, and subsequently, affect the quantity and function of neuronal cells. In addition, it appears that there is no clear initiator between microglia and the pathophysiological features such as tauopathy and Aβ deposition, and there are self-perpetuating cycles of inflammation.

 

Alzheimer’s disease

There is evidence to suggest that the immune system affects AD, an age-dependent progressive neurodegenerative disease that affects the CNS; however, further studies are needed to expand the connection (16)(36)(37).

AD is the leading cause of neurodegenerative dementia in the elderly, contributing to 60-70% of cases (38). The clinical presentation is characterized by progressive memory decline, impaired executive function, impairment in cognitive domains, and behavioral and psychological symptoms (39). The pathophysiological features of AD include neurofibrillary tangles (NFT) of intracellular tau protein aggregates, Aβ plaques, gliosis, and neurodegeneration (40)(41)(42).

A history of TBIs has been associated with dementia and is considered a risk factor for AD (43)(44). TBIs can lead to white matter, neuronal damage, p-tau, and Aβ deposition (45). Neuroinflammation occurs in AD by gliosis, the injury response in the CNS of activation and proliferation of microglia and astrocytes (39). Chronic neuroinflammation after TBI results in primed microglia, triggering inflammatory cascades causing neuronal damage, which may contribute to the onset and progression of AD (46, 47).

Although the process is still unclear, Aβ has neurotoxic and inflammatory effects that play an important role in the progression of AD (48). Data has shown that microglial activation may follow Aβ deposition in AD (40), but Aβ deposition may also be induced by activated microglia (49-51), so the cause and effect relationship is unclear. However, there is a close association between Aβ deposition and activated microglia. Aβ adheres to microglia, promoting synthesization and secretion of inflammatory mediators and progressing the disease. At the same time, activated microglia can phagocytize and clear Aβ plaques (48).

A similar association can be made for phosphorylated tau protein accumulation, another main hallmark of AD. Primed microglia and chronic neuroinflammation can boost tau protein and lead to the formation of NFT of intracellular tau protein aggregates (16). Human extracellular tau collection may be internalized by microglia, glial cells, and neurons may spread between cells and progress the disease (52). Concurrently, activated microglia also phagocytize phosphorylated tau protein and can release beneficial neurotrophic factors and antioxidants, limiting AD progression (53). The production of Aβ, phosphorylated tau, neuroinflammation, and neuronal damage caused by microglial activation is closely affiliated with AD pathogenesis (16).

 

Parkinson’s disease

PD is a progressive neurodegenerative disorder characterized by α-synuclein-containing Lewy bodies and the loss of dopaminergic neurons in the substantia nigra (SN). It is a prevalent movement disorder for older-aged adults, affecting 1% of those above 60 years of age (54). Clinical symptoms include resting tremors, impaired posture and balance, rigidity, and bradykinesia (55). Neuropathological features include the loss of dopaminergic neurons in the SN, intraneuronal inclusions called Lewy bodies, neuroinflammation, and gliosis (56). Dopaminergic neuron degeneration usually corresponds with the buildup of misfolded α-synuclein aggregates called Lewy bodies, which are spread throughout the SN and other brain regions (57). In addition, much evidence shows that microglial activation is a substantial pathological feature of PD and relative cognitive decline (58-64); however, microglia’s exact role is still unclear.

Postmortem PD brains have shown microglial activation and neuroimaging studies during disease development, and both the innate and adaptive immune systems have been implicated in PD. In 1988, McGeer et al. described microglia activation in PD when they discovered human leukocyte antigen DR (HLA-DR) expression in the SN of human brains (58). Further research showed TNF expression in SN (65) and raised levels of pro-inflammatory cytokines in the brain and cerebrospinal fluid (CSF) (66-68). In addition, and further supporting the involvement of neuroinflammation in PD, some non-steroidal anti-inflammatory drugs (NSAIDs) have been seen to have a protective effect on the incidence of PD in epidemiological studies (69-70).

Aging and chronic psychological stress are the two main environmental risk factors for PD, and they are also responsible for increasing pro-inflammatory mediators within the CNS and altering microglial functions. Microglial activation and the release of pro-inflammatory cytokines, including TNF, IL-6, IL-1β, and interferon-γ (INF-γ), could lead to neuronal loss (71) or could follow as a consequence, as the death of dopaminergic neurons can cause pro-inflammatory microglial “phagoptotic” phenotypes.

Microglia respond early to α-synuclein in neurons, correlated with MHCII expression, and contribute to PD by inflammation and phagocytosis of α-synuclein, factors associated with neurodegeneration (72). Microglia may be important for toxic α-synuclein clearance in neurons; however, microglial activation may also harm the neurons and lead to their death, which can be seen by decreased glucose metabolism in different brain regions of PD patients (73). a-synuclein aggregation causes neuronal dysfunction and death, but microgliosis can occur before neuronal death, not solely as a consequence (74). After neuron cell death, microglial activation is correlated with CD68 (75). Pro and anti-inflammatory functions of microglia can create imbalance, leading to chronic neuroinflammation that could lead to the onset and progression of PD (76).

               

Chronic traumatic encephalopathy

CTE is a progressive neurodegenerative disease believed to be caused by repeated episodes of mild TBI and is often found in athletes, such as boxers and football players, and military veterans who experienced combat. It is difficult to describe the prevalence of CTE, especially considering that diagnosis is made post-mortem by histopathological brain analysis. However, due to the widespread global practice and growing popularity of combat sports where concussive and subconcussive blows are common, added to the global number of military service members engaging in combat, considering that just among U.S. Military service members, approximately 430,000 TBIs were reported from 2000 to 2020 (77), it must be assumed that the incidence is extremely high. In a study of 85 post-mortem brains of athletes donated for research, 68 showed signs of CTE (78). Another recent and larger epidemiological study found that 6% of population-based brains showed CTE (79).

Clinical symptoms of CTE usually appear 8 to 10 years after repetitive mild TBI, and the patient may show aggression and irritability, impulsive behaviors, memory loss, and depression (80). As the disease progresses, dementia and parkinsonism may develop, and difficulties in speech and gait may present. It is a tauopathy characterized by hyperphosphorylated tau protein tangles, disseminated microgliosis, and astrocytosis (81). Some CTE cases, approximately 40%, showed Aβ plaques, which was age-dependent (82, 83).

Studies continue to indicate that chronic neuroinflammation can lead to CTE (6). Rodent studies of repetitive mild TBI have shown that following injury, neuroinflammation, and glial changes occur before tau protein pathology (84)(85). Neuroinflammation has been linked with higher tau pathology in CTE, contributing to its development and progression, with microglial cells playing a role (86). Microglial reactivity is a common feature of CTE, along with the accumulation of abnormal tau by astrocytes and neurons, where it is irregularly distributed within sulci (81)(87).

Repeated mild TBI can lead to chronic neuroinflammation and primed microglia, which release pro-inflammatory cytokine mediators. Studies have shown that increased density of CD68+ microglia in the brains of sports players was linked to CTE severity and that the time duration of repeated TBI was also associated with CD68 density (86). The chronic state of microglial activation may exacerbate tauopathy, initiating the formation of NFT and phosphorylated tau deposition, and tau can induce neuroinflammation in turn, as seen in rodent models, resulting in a self-perpetuating cycle of inflammation (88). However, it is still unclear which initiates the other, and further studies are needed to explore this relationship.

Future therapeutic implications

Neuroinflammation-based treatments for TBI are now being investigated, and with further clinical trials and research, targeting microglia activation could be beneficial. The first-stage inflammatory reaction following acute TBI is beneficial to help restore brain homeostasis. However, when chronically activated, primed microglia can harm the CNS, releasing pro-inflammatory molecules and causing secondary injury with neuronal damage that could lead to neurodegenerative pathologies. For this, medical management is targeted toward preventing the second-stage injury and limiting the harmful effects of primed microglia in chronic neuroinflammation.

Research advances are unveiling new findings in the study of post-TBI neuroinflammation, and new therapeutic targets are being explored. In addition, research is being done to determine the critical periods for treatment.

There is evidence showing the harmful effects of chronic neuroinflammation after TBI and the role of microglia during this secondary injury process. In some studies, microglial depletion was able to inhibit or stop this damage. Therefore, confining the microglial response to the acute, primary injury of TBI and limiting it during the neuroinflammatory secondary injury phase is a possible approach to limit neuroinflammation. This idea could be a potential therapeutic avenue for neuroprotection, although further research must be conducted (12).

Clinical trials treating TBI patients with anti-inflammatory drugs have not shown great success, and limited success has only been achieved using progesterone in younger patients (89). In addition, because of the large possible variation in outcome after TBI and the complexity of immune activation, successful treatment must be targeted to inflammatory mediators and interindividual differences such as age, genetic predisposition, and history of secondary injuries. Finally, it must be initiated at the correct time frame (89).

The manipulation of microglia activation and neuroimaging using PET and autoradiography are being used to identify the role that microglia play post-injury. In imaging, selective PET ligands for amyloid, tau, and neuroinflammation can provide insight into the post-TBI neurodegenerative process and help determine future therapeutic approaches (6). One possible treatment approach is manipulating microglia and activating them to become a beneficial phenotype rather than a destructive one (90).

Immunotherapy needs to be focused on the right moment, targeting the moment of positive immune activity to aid reparation and debris clearance and then decreasing the negative damage of chronic inflammation afterward.

One positive achievement in treatment is aerobic exercise, which has displayed the importance of intervention timing concerning benefits in neuroinflammation and neuroprotection. Post-TBI exercise intervention has shown overall cognitive benefits in mild to moderate TBI patients, with improvements in cognitive functioning and cardiorespiratory fitness (91). In a mouse study by Piao et al., the initiation of aerobic exercise after 5 weeks after moderate TBI promoted neurogenesis, benefited cognitive recovery, and attenuated the inflammatory response. In the same study, it was also seen that earlier initiation of exercise provided different results, with no cognitive benefits and a neurotoxic pro-inflammatory response, opposing the classically held view that neuroprotection can only be achieved with early intervention (92). However, previous training before TBI was also seen to provide a beneficial preventative effect on the cerebral inflammatory response following severe TBI, an important discovery that implicates exercise induces metabolic changes that can positively alter the long-term inflammation process following TBI and limit neuronal damage (93).

The take-away message from these exercise studies, and the overall evidence so far, is that there are different time frames for intervention during the inflammatory process following TBI, and interindividual differences and TBI history are important modifying factors accounted for.

CONCLUSION

 

TBI is a global source of death, disability, and morbidity and can have devastating long-term consequences for patients. It provokes an initial inflammatory response which can lead to secondary injury and chronic neuroinflammation with effects on cognitive functioning and the development of neurodegenerative diseases. In addition, a history of repeated TBI, even mild, puts a person at higher risk.

Microglia respond to acute injury after TBI and are beneficial for reparation. However, the continuous state of primed microglia that follows can be destructive and cause neuronal dysfunction.

In neurodegenerative diseases such as AD, PD, and CTE, characterized by abnormal accumulation of phosphorylated tau proteins and Aβ peptide deposits in the brain, neuroinflammation and microglial activation have been implicated in neuronal damage, disease worsening, and outcome.

Genetic and pharmacological manipulations of microglia activation are now being used to unveil their significance in post-TBI inflammation, and neuroimaging studies are being used to define the role of microglia. The timing seems to be of the utmost importance in therapy, as early or late intervention has different effects on immune function, dependent on many specific patient factors.

Because of the number of people affected by TBI worldwide and the correlation between neuroinflammation and neurodegenerative disease, it is highly important to continue research in this field to develop treatment options to improve patient outcomes.

Conflict of interest

The authors declare that they have no conflict of interest.

 

REFERENCES

 

  1. Centers for Disease Control. Intimate Partner Violence and Sexual Violence Prevention | CDC. www.cdc.gov. Published January 8, 2020. https://www.cdc.gov/injury/stateprograms/topic_traumatic-brain-injury.html#:~:text=Traumatic%20brain%20injury%20(TBI)%2C
  2. Centers for Disease Control. Surveillance Report of Traumatic Brain Injury-Related Emergency Department Visits, Hospitalizations, and Deaths UNITED STATES, 2014| CDC. www.cdc.gov. Accessed September 19, 2021. https://www.cdc.gov/traumaticbraininjury/pdf/TBI-Surveillance-Report-FINAL_508.pdf
  3. Faul M, Coronado V. Epidemiology of traumatic brain injury. Handbook of Clinical Neurology. 2015;127:3-13. doi:10.1016/b978-0-444-52892-6.00001-5
  4. Teasdale G, Jennett B. ASSESSMENT OF COMA AND IMPAIRED CONSCIOUSNESS: A Practical Scale. The Lancet. 1974;2(7872):81-84. doi:10.1016/s0140-6736(74)91871-6
  5. Corps KN, Roth TL, McGavern DB. Inflammation and Neuroprotection in Traumatic Brain Injury. JAMA Neurology. 2015;72(3):355. doi:10.1001/jamaneurol.2014.3558
  6. Simon DW, McGeachy MJ, Bayır H, Clark RSB, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nature Reviews Neurology. 2017;13(3):171-191. doi:10.1038/nrneurol.2017.13
  7. Salmond CH, Menon DK, Chatfield DA, Pickard JD, Sahakian BJ. Changes over time in cognitive and structural profiles of head injury survivors. Neuropsychologia. 2006;44(10):1995-1998. doi:10.1016/j.neuropsychologia.2006.03.013
  8. Himanen L, Portin R, Isoniemi H, Helenius H, Kurki T, Tenovuo O. Longitudinal cognitive changes in traumatic brain injury: A 30-year follow-up study. Neurology. 2006;66(2):187-192. doi:10.1212/01.wnl.0000194264.60150.d3
  9. Almeida OP, Hankey GJ, Yeap BB, Golledge J, Flicker L. Prevalence, associated factors, mood and cognitive outcomes of traumatic brain injury in later life: the health in men study (HIMS). International Journal of Geriatric Psychiatry. 2015;30(12):1215-1223. doi:10.1002/gps.4276
  10. Makinde HM, Just TB, Cuda CM, Perlman H, Schwulst SJ. The Role of Microglia in the Etiology and Evolution of Chronic Traumatic Encephalopathy. Shock (Augusta, Ga). 2017;48(3):276-283. doi:10.1097/SHK.0000000000000859
  11. Dixon KJ. Pathophysiology of Traumatic Brain Injury. Physical Medicine and Rehabilitation Clinics of North America. 2017;28(2):215-225. doi:10.1016/j.pmr.2016.12.001
  12. Bachstetter AD, Rowe RK, Kaneko M, Goulding D, Lifshitz J, Van Eldik LJ. The p38α MAPK regulates microglial responsiveness to diffuse traumatic brain injury. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2013;33(14):6143-6153. doi:10.1523/JNEUROSCI.5399-12.2013
  13. Postolache TT, Wadhawan A, Can A, et al. Inflammation in Traumatic Brain Injury. Journal of Alzheimer’s Disease. 2020;74(1):1-28. doi:10.3233/jad-191150
  14. Davalos D, Grutzendler J, Yang G, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nature Neuroscience. 2005;8(6):752-758. doi:10.1038/nn1472
  15. DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. Journal of Neurochemistry. 2016;139(Suppl 2):136-153. doi:10.1111/jnc.13607
  16. Li JW, Zong Y, Cao XP, Tan L, Tan L. Microglial priming in Alzheimer’s disease. Annals of Translational Medicine. 2018;6(10):176-176. doi:10.21037/atm.2018.04.22
  17. Muccigrosso MM, Ford J, Benner B, et al. Cognitive Deficits Develop 1 month after Diffuse Brain Injury and are Exaggerated by Microglia-Associated Reactivity to Peripheral Immune Challenge. Brain, behavior, and immunity. 2016;54:95-109. doi:10.1016/j.bbi.2016.01.009
  18. Ziebell JM, Taylor SE, Cao T, Harrison JL, Lifshitz J. Rod microglia: elongation, alignment, and coupling to form trains across the somatosensory cortex after experimental diffuse brain injury. Journal of Neuroinflammation. 2012;9(1). doi:10.1186/1742-2094-9-247
  19. Mouzon BC, Bachmeier C, Ferro A, et al. Chronic neuropathological and neurobehavioral changes in a repetitive mild traumatic brain injury model. Annals of Neurology. 2014;75(2):241-254. doi:10.1002/ana.24064
  20. Loane DJ, Kumar A, Stoica BA, Cabatbat R, Faden AI. Progressive Neurodegeneration After Experimental Brain Trauma. Journal of Neuropathology & Experimental Neurology. 2014;73(1):14-29. doi:10.1097/nen.0000000000000021
  21. Fenn AM, Gensel JC, Huang Y, Popovich PG, Lifshitz J, Godbout JP. Immune Activation Promotes Depression 1 Month After Diffuse Brain Injury: A Role for Primed Microglia. Biological Psychiatry. 2014;76(7):575-584. doi:10.1016/j.biopsych.2013.10.014
  22. Ramlackhansingh AF, Brooks DJ, Greenwood RJ, et al. Inflammation after trauma: Microglial activation and traumatic brain injury. Annals of Neurology. 2011;70(3):374-383. doi:10.1002/ana.22455
  23. Witcher KG, Bray CE, Chunchai T, et al. Traumatic Brain Injury Causes Chronic Cortical Inflammation and Neuronal Dysfunction Mediated by Microglia. The Journal of Neuroscience. 2021;41(7):1597-1616. doi:10.1523/jneurosci.2469-20.2020
  24. 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 Neurology. 2017;74(1):67. doi:10.1001/jamaneurol.2016.3764
  25. Juengst SB, Kumar RG, Arenth PM, Wagner AK. Exploratory associations with Tumor Necrosis Factor-α, disinhibition and suicidal endorsement after traumatic brain injury. Brain, Behavior, and Immunity. 2014;41:134-143. doi:10.1016/j.bbi.2014.05.020
  26. Gupta J, Nebreda AR. Roles of p38α mitogen‐activated protein kinase in mouse models of inflammatory diseases and cancer. The FEBS Journal. 2015;282(10):1841-1857. doi:10.1111/febs.13250
  27. Raivich G, Bohatschek M, Kloss CUA, Werner A, Jones LL, Kreutzberg GW. Neuroglial activation repertoire in the injured brain: graded response, molecular mechanisms and cues to physiological function. Brain Research Reviews. 1999;30(1):77-105. doi:10.1016/s0165-0173(99)00007-7
  28. Delpech JC, Madore C, Nadjar A, Joffre C, Wohleb ES, Layé S. Microglia in neuronal plasticity: Influence of stress. Neuropharmacology. 2015;96(Pt A):19-28. doi:10.1016/j.neuropharm.2014.12.034
  29. Gorse KM, Lafrenaye AD. The Importance of Inter-Species Variation in Traumatic Brain Injury-Induced Alterations of Microglial-Axonal Interactions. Frontiers in Neurology. 2018;9. doi:10.3389/fneur.2018.00778
  30. Wofford KL, Harris JP, Browne KD, et al. Rapid neuroinflammatory response localized to injured neurons after diffuse traumatic brain injury in swine. Experimental Neurology. 2017;290:85-94. doi:10.1016/j.expneurol.2017.01.004
  31. Hellewell SC, Yan EB, Agyapomaa DA, Bye N, Morganti-Kossmann MC. Post-Traumatic Hypoxia Exacerbates Brain Tissue Damage: Analysis of Axonal Injury and Glial Responses. Journal of Neurotrauma. 2010;27(11):1997-2010. doi:10.1089/neu.2009.1245
  32. Semple BD, Bye N, Rancan M, Ziebell JM, Morganti-Kossmann MC. Role of CCL2 (MCP-1) in Traumatic Brain Injury (TBI): Evidence from Severe TBI Patients and CCL2−/− Mice. Journal of Cerebral Blood Flow & Metabolism. 2009;30(4):769-782. doi:10.1038/jcbfm.2009.262
  33. Das M, Leonardo CC, Rangooni S, Pennypacker KR, Mohapatra S, Mohapatra SS. Lateral fluid percussion injury of the brain induces CCL20 inflammatory chemokine expression in rats. Journal of Neuroinflammation. 2011;8(1). doi:10.1186/1742-2094-8-148
  34. Rapoport MJ. Depression Following Traumatic Brain Injury. CNS Drugs. 2012;26(2):111-121. doi:10.2165/11599560-000000000-00000
  35. Chauhan NB. Chronic neurodegenerative consequences of traumatic brain injury. Restorative Neurology and Neuroscience. 2014;32(2):337-365. doi:10.3233/rnn-130354
  36. Heyman A, Wilkinson WE, Stafford JA, Helms MJ, Sigmon AH, Weinberg T. Alzheimer’s disease: A study of epidemiological aspects. Annals of Neurology. 1984;15(4):335-341. doi:10.1002/ana.410150406
  37. Plassman BL, Havlik RJ, Steffens DC, et al. Documented head injury in early adulthood and risk of Alzheimer’s disease and other dementias. Neurology. 2000;55(8):1158-1166. doi:10.1212/wnl.55.8.1158
  38. World Health Organization. Dementia. Who.int. Published September 2, 2021. https://www.who.int/news-room/fact-sheets/detail/dementia
  39. Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Molecular Neurodegeneration. 2020;15(1). doi:10.1186/s13024-020-00391-7
  40. Itagaki S, McGeer P, Akiyama H, Zhu S, Selkoe D. Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease. Journal of Neuroimmunology. 1989;24(3):173-182. doi:10.1016/0165-5728(89)90115-x
  41. Terry RD, Masliah E, Salmon DP, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Annals of neurology. 1991;30(4):572-580. doi:10.1002/ana.410300410
  42. Iqbal K, Grundke-Iqbal I. Neurofibrillary pathology leads to synaptic loss and not the other way around in Alzheimer disease. Journal of Alzheimer’s Disease. 2002;4(3):235-238. doi:10.3233/jad-2002-4313
  43. Mendez MF. What is the Relationship of Traumatic Brain Injury to Dementia? Journal of Alzheimer’s Disease. 2017;57(3):667-681. doi:10.3233/jad-161002
  44. Nordström A, Nordström P. Traumatic brain injury and the risk of dementia diagnosis: A nationwide cohort study. Menon D, ed. PLOS Medicine. 2018;15(1):e1002496. doi:10.1371/journal.pmed.1002496
  45. Bloom GS. Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA neurology. 2014;71(4):505-508. doi:10.1001/jamaneurol.2013.5847
  46. Perry VH, Nicoll JAR, Holmes C. Microglia in neurodegenerative disease. Nature Reviews Neurology. 2010;6(4):193-201. doi:10.1038/nrneurol.2010.17
  47. Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med. 2015;3:136. doi:10.3978/j.issn.2305-5839.2015.03.49
  48. Yu Y, Ye RD. Microglial Aβ Receptors in Alzheimer’s Disease. Cellular and Molecular Neurobiology. 2014;35(1):71-83. doi:10.1007/s10571-014-0101-6
  49. Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends in Immunology. 2007;28(3):138-145. doi:10.1016/j.it.2007.01.005
  50. Von Bernhardi R. Glial cell dysregulation: a new perspective on Alzheimer disease. Neurotoxicity Research. 2007;12(4):215-232. doi:10.1007/bf03033906
  51. Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2012;493(7434):674-678. doi:10.1038/nature11729
  52. Dujardin S, Lécolle K, Caillierez R, et al. Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies. Acta Neuropathologica Communications. 2014;2(1). doi:10.1186/2051-5960-2-14
  53. Dudvarski Stankovic N, Teodorczyk M, Ploen R, Zipp F, Schmidt MHH. Microglia–blood vessel interactions: a double-edged sword in brain pathologies. Acta Neuropathologica. 2015;131(3):347-363. doi:10.1007/s00401-015-1524-y
  54. Tysnes OB, Storstein A. Epidemiology of Parkinson’s disease. Journal of Neural Transmission. 2017;124(8):901-905. doi:10.1007/s00702-017-1686-y
  55. Jankovic J. Parkinson’s disease: clinical features and diagnosis. Journal of Neurology, Neurosurgery & Psychiatry. 2008;79(4):368-376. doi:10.1136/jnnp.2007.131045
  56. Tansey MG, Romero‐Ramos M. Immune system responses in Parkinson’s disease: Early and dynamic. European Journal of Neuroscience. 2019;49(3):364-383. doi:10.1111/ejn.14290
  57. Tansey MG, Goldberg MS. Neuroinflammation in Parkinson’s disease: Its role in neuronal death and implications for therapeutic intervention. Neurobiology of Disease. 2010;37(3):510-518. doi:10.1016/j.nbd.2009.11.004
  58. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38(8):1285-1285. doi:10.1212/wnl.38.8.1285
  59. Vawter MP, Dillon-Carter O, Tourtellotte WW, Carvey P, Freed WJ. TGFβ1 and TGFβ2 Concentrations Are Elevated in Parkinson’s Disease in Ventricular Cerebrospinal Fluid. Experimental Neurology. 1996;142(2):313-322. doi:10.1006/exnr.1996.0200
  60. Gao HM, Jiang J, Wilson B, Zhang W, Hong JS, Liu B. Microglial activation-mediated delayed and progressive degeneration of rat nigral dopaminergic neurons: relevance to Parkinson’s disease. Journal of Neurochemistry. 2002;81(6):1285-1297. doi:10.1046/j.1471-4159.2002.00928.x
  61. Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, Hashizume Y. Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson’s disease brains. Acta Neuropathologica. 2003;106(6):518-526. doi:10.1007/s00401-003-0766-2
  62. Mount MP, Lira A, Grimes D, et al. Involvement of Interferon- in Microglial-Mediated Loss of Dopaminergic Neurons. Journal of Neuroscience. 2007;27(12):3328-3337. doi:10.1523/jneurosci.5321-06.2007
  63. Litteljohn D, Hayley S. Cytokines as Potential Biomarkers for Parkinson’s Disease: A Multiplex Approach. Methods in Molecular Biology. 2012;934:121-144. doi:10.1007/978-1-62703-071-7_7
  64. Doorn KJ, Moors T, Drukarch B, van de Berg WD, Lucassen PJ, van Dam AM. Microglial phenotypes and toll-like receptor 2 in the substantia nigra and hippocampus of incidental Lewy body disease cases and Parkinson’s disease patients. Acta Neuropathologica Communications. 2014;2(1). doi:10.1186/s40478-014-0090-1
  65. Boka G, Anglade P, Wallach D, Javoy-Agid F, Agid Y, Hirsch EC. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson’s disease. Neuroscience Letters. 1994;172(1-2):151-154. doi:10.1016/0304-3940(94)90684-x
  66. Mogi M, Harada M, Kondo T, et al. Interleukin-1β, interleukin-6, epidermal growth factor and transforming growth factor-α are elevated in the brain from parkinsonian patients. Neuroscience Letters. 1994;180(2):147-150. doi:10.1016/0304-3940(94)90508-8
  67. Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T. Tumor necrosis factor-α (TNF-α) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neuroscience Letters. 1994;165(1-2):208-210. doi:10.1016/0304-3940(94)90746-3
  68. Mogi M, Harada M, Kondo T, Narabayashi H, Riederer P, Nagatsu T. Transforming growth factor-β1 levels are elevated in the striatum and in ventricular cerebrospinal fluid in Parkinson’s disease. Neuroscience Letters. 1995;193(2):129-132. doi:10.1016/0304-3940(95)11686-q
  69. Gagne JJ, Power MC. Anti-inflammatory drugs and risk of Parkinson disease: A meta-analysis. Neurology. 2010;74(12):995-1002. doi:10.1212/wnl.0b013e3181d5a4a3
  70. Gao X, Chen H, Schwarzschild MA, Ascherio A. Use of ibuprofen and risk of Parkinson disease. Neurology. 2011;76(10):863-869. doi:10.1212/wnl.0b013e31820f2d79
  71. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Progress in Neurobiology. 2005;76(2):77-98. doi:10.1016/j.pneurobio.2005.06.004
  72. Ho MS. Microglia in Parkinson’s Disease. Advances in Experimental Medicine and Biology. 2019;1175:335-353. doi:10.1007/978-981-13-9913-8_13
  73. Fan Z, Aman Y, Ahmed I, et al. Influence of microglial activation on neuronal function in Alzheimer’s and Parkinson’s disease dementia. Alzheimer’s & Dementia. 2014;11(6):608-621.e7. doi:10.1016/j.jalz.2014.06.016
  74. Watson MB, Richter F, Lee SK, et al. Regionally-specific microglial activation in young mice over-expressing human wildtype alpha-synuclein. Experimental Neurology. 2012;237(2):318-334. doi:10.1016/j.expneurol.2012.06.025
  75. Sanchez-Guajardo V, Febbraro F, Kirik D, Romero-Ramos M. Microglia Acquire Distinct Activation Profiles Depending on the Degree of α-Synuclein Neuropathology in a rAAV Based Model of Parkinson’s Disease. Cookson MR, ed. PLoS ONE. 2010;5(1):e8784. doi:10.1371/journal.pone.0008784
  76. Lecours C, Bordeleau M, Cantin L, Parent M, Paolo TD, Tremblay MÈ. Microglial Implication in Parkinson’s Disease: Loss of Beneficial Physiological Roles or Gain of Inflammatory Functions? Frontiers in Cellular Neuroscience. 2018;12:282. doi:10.3389/fncel.2018.00282
  77. Centers for Disease Control. TBI Among Service Members and Veterans | Concussion | Traumatic Brain Injury | CDC Injury Center. www.cdc.gov. Published May 12, 2021. Accessed September 20, 2021. https://www.cdc.gov/traumaticbraininjury/military/index.html
  78. McKee AC, Stein TD, Nowinski CJ, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain. 2012;136(1):43-64. doi:10.1093/brain/aws307
  79. Bieniek KF, Blessing MM, Heckman MG, et al. Association between contact sports participation and chronic traumatic encephalopathy: a retrospective cohort study. Brain Pathology. 2019;30(1):63-74. doi:10.1111/bpa.12757
  80. McKee AC, Cantu RC, Nowinski CJ, et al. Chronic Traumatic Encephalopathy in Athletes: Progressive Tauopathy After Repetitive Head Injury. Journal of Neuropathology & Experimental Neurology. 2009;68(7):709-735. doi:10.1097/nen.0b013e3181a9d503
  81. Goldstein LE, Fisher AM, Tagge CA, et al. Chronic Traumatic Encephalopathy in Blast-Exposed Military Veterans and a Blast Neurotrauma Mouse Model. Science Translational Medicine. 2012;4(134):134ra60-134ra60. doi:10.1126/scitranslmed.3003716
  82. McKee AC, Stein TD, Kiernan PT, Alvarez VE. The Neuropathology of Chronic Traumatic Encephalopathy. Brain Pathology. 2015;25(3):350-364. doi:10.1111/bpa.12248
  83. Edwards G, Moreno-Gonzalez I, Soto C. Amyloid-beta and tau pathology following repetitive mild traumatic brain injury. Biochemical and Biophysical Research Communications. 2017;483(4):1137-1142. doi:10.1016/j.bbrc.2016.07.123
  84. Petraglia A, Plog B, Dayawansa S, et al. The pathophysiology underlying repetitive mild traumatic brain injury in a novel mouse model of chronic traumatic encephalopathy. Surgical Neurology International. 2014;5(1):184. doi:10.4103/2152-7806.147566
  85. 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. doi:10.1038/nature14658
  86. Cherry JD, Tripodis Y, Alvarez VE, et al. Microglial neuroinflammation contributes to tau accumulation in chronic traumatic encephalopathy. Acta Neuropathologica Communications. 2016;4(1). doi:10.1186/s40478-016-0382-8
  87. McKee AC, Cairns NJ, Dickson DW, et al. The first NINDS/NIBIB consensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathologica. 2015;131(1):75-86. doi:10.1007/s00401-015-1515-z
  88. Maphis N, Xu G, Kokiko-Cochran ON, et al. loss of tau rescues inflammation-mediated neurodegeneration. Frontiers in Neuroscience. 2015;9:196. doi:10.3389/fnins.2015.00196
  89. Begemann M, Leon M, van der Horn HJ, van der Naalt J, Sommer I. Drugs with anti-inflammatory effects to improve outcome of traumatic brain injury: a meta-analysis. Scientific Reports. 2020;10(1). doi:10.1038/s41598-020-73227-5
  90. Donat CK, Scott G, Gentleman SM, Sastre M. Microglial Activation in Traumatic Brain Injury. Frontiers in Aging Neuroscience. 2017;9:208. doi:10.3389/fnagi.2017.00208
  91. Chin LM, Keyser RE, Dsurney J, Chan L. Improved Cognitive Performance Following Aerobic Exercise Training in People With Traumatic Brain Injury. Archives of Physical Medicine and Rehabilitation. 2015;96(4):754-759. doi:10.1016/j.apmr.2014.11.009
  92. Piao CS, Stoica BA, Wu J, et al. Late exercise reduces neuroinflammation and cognitive dysfunction after traumatic brain injury. Neurobiology of Disease. 2013;54:252-263. doi:10.1016/j.nbd.2012.12.017
  93. de Castro MRT, Ferreira AP de O, Busanello GL, et al. Previous physical exercise alters the hepatic profile of oxidative-inflammatory status and limits the secondary brain damage induced by severe traumatic brain injury in rats. The Journal of Physiology. 2017;595(17):6023-6044. doi:10.1113/jp273933

You may also like...