Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 14.
Published in final edited form as: Nat Rev Neurol. 2024 Apr 3;20(5):298–312. doi: 10.1038/s41582-024-00954-y

Insights into epileptogenesis from post-traumatic epilepsy

Matthew Pease 1,, Kunal Gupta 2, Solomon L Moshé 3,4,5, Daniel J Correa 3, Aristea S Galanopoulou 3,4, David O Okonkwo 6, Jorge Gonzalez-Martinez 6, Lori Shutter 6,7,8, Ramon Diaz-Arrastia 9, James F Castellano 8
PMCID: PMC12257072  NIHMSID: NIHMS2095366  PMID: 38570704

Abstract

Post-traumatic epilepsy (PTE) accounts for 5% of all epilepsies. The incidence of PTE after traumatic brain injury (TBI) depends on the severity of injury, approaching one in three in groups with the most severe injuries. The repeated seizures that characterize PTE impair neurological recovery and increase the risk of poor outcomes after TBI. Given this high risk of recurrent seizures and the relatively short latency period for their development after injury, PTE serves as a model disease to understand human epileptogenesis and trial novel anti-epileptogenic therapies. Epileptogenesis is the process whereby previously normal brain tissue becomes prone to recurrent abnormal electrical activity, ultimately resulting in seizures. In this Review, we describe the clinical course of PTE and highlight promising research into epileptogenesis and treatment using animal models of PTE. Clinical, imaging, EEG and fluid biomarkers are being developed to aid the identification of patients at high risk of PTE who might benefit from anti-epileptogenic therapies. Studies in preclinical models of PTE have identified tractable pathways and novel therapeutic strategies that can potentially prevent epilepsy, which remain to be validated in humans. In addition to improving outcomes after TBI, advances in PTE research are likely to provide therapeutic insights that are relevant to all epilepsies.

Introduction

Traumatic injuries account for more lost years and productivity than cancer or cardiovascular disease1. Affecting 55 million people worldwide, traumatic brain injury (TBI) is a leading cause of death and disability2,3. Post-traumatic epilepsy (PTE) is a major driver of the neurological, emotional and occupational disability associated with TBI. Seizures cause secondary injury after TBI, often exacerbating neurodegeneration, inflammation and blood–brain barrier (BBB) dysfunction4,5. PTE impairs neurological recovery after TBI and is independently associated with poor functional outcomes6,7. PTE affects up to one-third of individuals with severe TBI and accounts for 20% of structural epilepsies overall, so effective treatment of this condition provides an opportunity to reduce the burden of disability after TBI8,9.

PTE develops through epileptogenesis1012 — a chronic process whereby normally functioning brain tissue becomes prone to recurrent abnormal electrical activity that results in seizures. This continuous and prolonged process is traditionally thought to occur in three, often overlapping, clinical phases: first, an initial insult such as TBI; second, a latency period lasting months to years during which epileptic circuits develop; and third, the symptomatic phase with spontaneous, unprovoked seizures defining epilepsy13. Once epilepsy develops, epileptogenesis continues as seizures reinforce and expand epileptic circuits, potentially developing into drug-resistant epilepsy. Despite substantial progress in the development of over 30 anti-seizure medications (ASMs) over the past few decades, we still lack the ability to modify or alter the process of epileptogenesis in humans12,14,15.

The development of disease-modifying anti-epileptogenic medications (AEMs) is recognized as a public health priority by many national and international organizations, including the WHO, the NIH and the European Commission1618. PTE is an excellent model disease to elucidate the mechanisms of epileptogenesis and trial AEMs, as the initial trigger (TBI) is common and clearly defined, and epileptogenesis typically occurs over a relatively short latency period. Epileptogenesis continues over the disease course, as patients with PTE have higher rates of drug resistance than do individuals with other epilepsy types19,20. Multi-institutional consortia are developing clinical and preclinical trial networks to identify targeted therapies to better treat PTE2126.

In this Review, we first provide a clinical overview of PTE, describing its clinical characteristics, course and current treatment strategies. We then review epileptogenic mechanisms in PTE and tractable targets for AEMs. A major challenge in navigating the PTE research field is the range of different definitions for common terms. We define PTE as a single late (more than 7 days post-injury) post-traumatic seizure6,11 (PTS). The International League Against Epilepsy (ILAE) defines epilepsy as two seizures at least 24 h apart or a single seizure with a high risk of recurrence27 (more than 60% at 10 years). Owing to the high risk of seizure recurrence after TBI, a single late PTS (LPTS) meets the ILAE definition of PTE11,2830.

Epidemiology of PTE

Despite considerable efforts, the incidence of PTE has been difficult to define owing to inadequacies in seizure reporting and long-term follow-up and variations in study approaches. The available data indicate that PTE is common, with a prevalence of 50% among war veterans with penetrating TBI and around 33% among civilians with severe TBI20. Worldwide, PTE accounts for 5% of the 70 million cases of epilepsy worldwide and 20% of symptomatic epilepsies with a structural aetiology8,31,32. In a seminal study, the Rochester Epidemiology Project at the Mayo Clinic retrospectively studied PTE rates in patients with TBI from the 1930s to the 1980s10. This study established two guiding principles for PTE epidemiology: first, the rate of PTE rises considerably with increasing TBI severity; and second, PTE can occur years to decades after injury.

In the light of these findings, subsequent studies used prospective databases to better define the rate of PTE after TBI (Fig. 1). When the Glasgow Coma Scale (GCS) was used to stratify injury severity, the rates of PTE after 1–2 years of follow-up were 12–26% for severe TBI7,29,33,34, 5–24% for moderate TBI7,33,34 and 1–8% for mild TBI7,34. Studies using the Abbreviated Injury Scale found similar results, with PTE rates ranging from 8% to 21% in people with moderate-to-severe TBI35,36. The main strength of these studies was the ability to clearly define ranges for the risk of PTE up to 2 years after injury. The prohibitive cost of prospectively monitoring large cohorts of patients for years after their injuries limits our understanding of how PTE evolves over long time periods. Similarly, these prospective studies are limited by loss to follow-up, with some studies only having seizure data available for 15–50% of patients7,36. The definition of seizures also varied, with many studies using non-validated epilepsy questionnaires7,36.

Fig. 1 |. Prevalence of post-traumatic epilepsy after moderate or severe traumatic brain injury.

Fig. 1 |

The graph shows the prevalence of post-traumatic epilepsy (PTE) over time after traumatic brain injury (TBI) for selected representative cohorts. Burke et al.7, Englander et al.34, Temkin et al.85, Ritter et al.29 and Tubi et al.47 prospectively recorded seizures using physician visits, patient forms or phone calls. Pease et al.11 recorded seizures through retrospective chart review of a prospectively maintained database, whereas DeGrauw et al.39 used insurance claims data. The data indicate that the PTE onset occurs during the first year post-TBI in most cases, although the risk of developing PTE persists for at least 5 years.

Another approach to tracking PTE incidence involved the use of large population-based electronic medical record systems10,35,37,38 or claims databases39. These studies had several important methodological differences from the prospective studies described earlier. First, many reported relative rather than absolute risk of developing PTE compared with matched controls37,38. Second, none of these studies used the GCS to stratify injury severity, instead relying on various inconsistent definitions. In a seminal study of PTE risk40, Annengers et al. found a hazard ratio of 17 for severe TBI (defined by the authors as brain contusion or loss of consciousness for >24 h), 2.9 for moderate TBI (defined as skull fracture or loss of consciousness or amnesia for >30 min) and 1.5 for mild TBI (defined as loss of consciousness or amnesia for <30 min). Finally, most of the studies used diagnostic codes or claims databases rather than patient-reported data to identify seizures37,38,41. Cumulatively, these studies showed that the risk of PTE increased with TBI severity, with intracerebral contusions conferring the greatest risk. The rates of PTE after 1 year tended to be lower than that in prospective studies: typically <5% overall and <10% even in people with severe TBI35,38,39,41. This discrepancy might reflect the difficulties of using medical coding or billing to identify seizures42,43, and also the likelihood that PTE is underdiagnosed and undertreated in community settings.

Mild TBI with a negative CT head scan is 10 times more common than moderate or severe TBI, affecting nearly one million people in North America yearly44, and is associated with the lowest rates of PTE post-injury. Although some older studies found that rates of PTE after this type of TBI were comparable to the annual incidence of epilepsy in the general population10,40 (60 in 10,000 individuals), more recent studies have found an increased risk, albeit small37,38,45. Repeated mild TBI also seems to increase the risk of epilepsy46.

Several studies, mainly retrospective, have determined that PTE can occur up to four decades after the inciting TBI10,11,20,29,39,41,47. A decade after TBI, the cumulative incidence of PTE in modern cohorts was 32–53% in people with more severe injuries11,47,48. The increased risk continues across the spectrum of TBI severity, with patients who sustained a concussion having double the relative risk at a decade post-injury compared with the general population10,39,41. Seizures that occur years to decades after injury seem to be similar to those occurring within the first year post-injury in terms of semiology and rates of treatment resistance20. Despite the long tail of PTE risk, PTE onset typically occurs early after TBI, often within the first 6–12 months6,10,34,41. This knowledge can inform the timing of PTE screening, with benefits not only for clinical management but also for clinical trial design.

A hallmark of PTE is its relative severity, including high seizure frequency and recurrence rates. The recurrence rates approach 80% in the first few years after index seizure11,2830, and almost half of the patients with PTE have five or more repeat seizures30. The rates of medication-resistant PTE range from 23% to 53%, compared with 30–40% for non-traumatic epilepsies11,17,20,30.

PTE is understudied in World Bank-designated low-and-middle-income countries, with many groups reporting incidences of 10% or less even in people with more severe injuries28,4952. This phenomenon probably reflects a combination of the difficulties of running expensive prospective studies in regions with limited resources, undertreatment and lack of surveillance for PTE. Nearly 80% of patients with epilepsy in low-income countries do not have access to treatment53. The high rate (15%) of PTE in epilepsy clinics in low-income countries, which is similar to the prevalence of symptomatic epilepsy in high-income countries, suggests undertreatment rather than reduced prevalence.

Risk factors for PTE

The main driver of PTE risk is injury severity, typically measured by the GCS and proxy measures of brain damage. However, the GCS is an imperfect measure of injury severity; for example, it has a loose correlation with mortality54, and prognostic models incorporating the GCS account for less than one-third of the variation in outcomes55,56. In multivariate prediction models for TBI, the GCS is frequently replaced by other markers of injury severity, including duration of post-traumatic amnesia57,58, midline shift59 and neurosurgical procedures11,58,60. Decompressive craniectomy, whereby nearly one-third of the calvarium of the skull is removed to accommodate swelling, is associated with an odds ratio of more than 4 for PTE in multivariate models, although this factor probably correlates with injury severity rather than being causative11,58.

Age correlates inversely with PTE risk, with younger adults at greater risk than older adults11,29. This finding could reflect the fact that younger patients are more likely to survive severe injuries and go on to develop PTE or, alternatively, that they have greater CNS plasticity, which increases susceptibility to the formation of epileptic circuits. Age is a major predictor of survival in most clinical prognostic models6163.

Most markers of injury severity correlate with underlying brain damage, which can be causative for PTE but is challenging to quantify64,65. For example, healing and gliosis after focal cortical injury could induce epileptogenesis by promoting the development of abnormal electric circuits in the cortex66. These circuits are reinforced into a network, thereby maintaining aberrant hypersynchronized electrical activity and causing seizures. One study showed that veterans with penetrating brain injuries, which often result in focal cortical injury, had more than a 50% lifetime risk of developing PTE48. Individuals with depressed skull fractures, who often have underlying cortical damage, also have a clinically increased risk of PTE59,67,68.

Deeper focal injuries also pose a risk of PTE64. Subcortical contusions, which also lead to abnormal healing and gliosis, are associated with an increased PTE risk33,69. Greater numbers and sizes of brain lesions, as well as increased surrounding oedema, increase PTE risk in a volume-dependent manner58,64,70. Incomplete gliosis surrounding focal lesions independently increases PTE risk, as healing occurs in a haphazard, abnormal pattern71. Studies from specialized epilepsy surgery clinics have corroborated the relationship between focal injuries and PTE. Patients in these clinics, who typically have long-standing PTE, predominantly have focal epilepsy, with almost one-half having a cortical focus72. As in the case of other focal epilepsies, temporal lobe seizures are the most common, followed by frontal lobe seizures; parietal and occipital foci are relatively uncommon72. This finding is probably attributable not only to the high prevalence of temporal lobe damage in TBI but also to the underlying differential epileptogenic potential of various brain regions. Large mass-occupying lesions that deform the brain parenchyma, such as subdural haematomas, are associated with underlying injury and greatly increased risks of PTE, with or without surgery11,59,60,73. By contrast, epidural haematomas, typically from skull fractures and laceration of the meningeal vessels, do not seem to increase PTE risk11,59.

Diffuse or extra-axial injuries also contribute to PTE. Diffuse axonal injury results from shear stress along white matter tracts. Damage to these tracts can persist for years and is prominent in patients with cognitive decline after TBI72. These diffuse injuries can modify white matter circuitry through Wallerian degeneration and sprouting of new aberrant, potentially epileptogenic, connections64,74. In addition, diffuse injuries increase PTE risk by damaging key epileptogenic brain regions such as the hippocampus75 or thalamus76, which modulate seizure suppression through network changes. Measuring diffuse injuries in the acute phase after TBI can be challenging.

Secondary brain injuries further exacerbate PTE risk. In the acute phase, elevations in intracranial pressure can cause widespread damage to white matter tracts, as well as neuroinflammation and neurodegeneration77,78. In the longer term, intracranial infections can also increase the risk of PTE11,79. Compared with a single TBI, repeated TBI months to years after the initial injury nearly doubles the PTE risk46.

The risk factors that affect PTE latency — that is, the time from injury to first seizure onset — are largely unknown. As we highlighted earlier, epidemiological studies have demonstrated that PTE onset can occur decades after injury29,33,39, although it typically manifests within the first few months up to a year after injury10,11,34 (Fig. 1). Only one study has evaluated risk factors for reduced PTE latency and found that age, injury severity and residual disability were associated with decreased time to PTE onset. Risk of seizure recurrence, although known to be high11,29,30, has not been studied in detail, but might be reduced by cerebrospinal fluid shunting11.

Subpopulations of people with PTE, such as those stratified by gender and socioeconomic status, have not been adequately studied. Males seem to have a higher risk of PTE and seizure recurrence than do females, although one group found that this phenomenon only applied to individuals with severe TBI46,59,80. Groups with disadvantaged social support networks, such as prisoners or those with alcohol misuse, are also at increased risk of PTE46,59,80. Race does not seem to influence PTE risk, although this has not been studied thoroughly29.

Early PTSs and PTE

PTSs are divided into early PTSs (EPTSs), which manifest within 7 days of trauma, and LPTSs, which manifest beyond 7 days11. EPTSs are considered to be provoked seizures, whereas LPTSs are unprovoked and are synonymous with PTE. EPTS rates vary depending on the injury severity, ranging from 0% to 4% for mild TBI and up to 15% without prophylaxis after severe TBI8185. EPTSs are associated with increased length of hospital stay and with non-home discharge and mortality36.

Some groups have further subdivided EPTSs to include an immediate seizure subgroup86. Although definitions vary, immediate seizures typically occur in the pre-hospital setting or before resuscitation. Early trauma research indicated that immediate PTSs did not confer an increased risk of PTE, as they were typically acutely provoked by injury rather than by the development of epileptogenic circuits in the brain29,40,87. Differentiating between seizures and pathological posturing responses can also be challenging in the pre-hospital setting. Some researchers have classified immediate EPTSs as any seizure within the first 24 h after TBI, often for the sake of convenience in large coding-based databases, in which it can be difficult to distinguish between seizure onset in pre-resuscitation and post-resuscitation settings29,88. Considering that most EPTSs occur during the first day85,89, this approach might inappropriately increase the risk attributed to immediate seizures and explain why one group found that immediate EPTSs increased PTE risk, contrary to other groups29.

Randomized controlled trials (RCTs), meta-analyses and guidelines indicate that ASM prophylaxis protects against EPTSs in people with severe TBI85,9092, providing an absolute risk reduction of nearly 11% (ref. 85). Clinical trials routinely measure ASM levels in the peripheral blood and adjust the dose as appropriate, although this is rarely done in clinical practice, which could affect real-world clinical outcomes93,94. All types of ASMs seem to be equally effective, although sodium valproate use increased mortality in one study95; most practitioners use levetiracetam or phenytoin92,96,97. Owing to the comatose nature of patients with severe TBI and their risk of electrographic seizures, guidelines recommend continuous EEG36,94,98,99.

The robust effectiveness of ASM prophylaxis in severe TBI has not been reproduced in people with mild or moderate TBI83,88,100102, possibly because many studies are underpowered to find an effect owing to lower EPTS rates and/or smaller absolute risk reduction in mild and moderate TBI. The benefits of ASM prophylaxis in mild and moderate TBI should be weighed against the risks of inappropriate continuation of ASM prophylaxis82,103, including infection104, agitation105, hyponatraemia106 and poor functional outcomes103.

EPTSs could provide important insights into epileptogenesis. Although the data are inconsistent, EPTS seems to confer an increased risk of PTE11,28,34,35,39,47,52,107, possibly through the early development of abnormal epileptogenic circuits. Patients with EPTSs have a shorter time to onset of PTE than those with LPTSs, although this association might be spurious as both EPTS and PTE latency are influenced by injury severity80. In contrast to people with EPTSs, ASM prophylaxis does not reduce the long-term risk of PTE85,92, probably reflecting the failure of current ASMs to modify or ameliorate epileptogenesis108. ASMs are seizure-management tools, and modern ASMs have similar efficacy to older generations of ASMs17,108. Although we cannot reliably modify epileptogenesis with ASMs at present, management of EPTSs might have a role in preventing PTE development — a possibility that should be explored in future studies.

PTE and long-term TBI outcomes

Whether PTE results in worsened disability after TBI, independently of injury severity, remains unproven, possibly owing to the large confounding effect of injury severity on both PTE risk and long-term outcomes. PTE is associated with worse outcomes when global measures, such as the Glasgow Outcome Scale (GOS) or the Extended GOS (GOSE)6,7,47, functional independence measures103,109,110, employment19, mental health111113 and cognitive decline114, are considered. However, it is difficult to fully account for the effects of PTE independently of injury severity, as markers of injury severity, such as GCS scores and pupil reactivity, have similar associations. Nonetheless, there are several mechanistic reasons why seizures might worsen neurological outcomes. PTE acts synergistically with injury severity, causing a ‘second hit’. Seizures worsen neural inflammation, which can promote neuronal cell death and remodelling of cognitive circuits, thereby limiting recovery from brain injury and exacerbating functional decline115,116. In addition, seizures can promote BBB breakdown, which can result in neurodegeneration.

Identifying the relative contribution of PTE to poor outcomes after TBI is challenging. Two studies controlled for injury severity using propensity matching and regression, and they found that PTE was an independent risk factor for poor functional outcomes based on GOS scores6,7. These approaches are limited, however, by difficulties in defining injury severity in TBI. Both large-scale, regression-based clinical prognostic models and advanced deep learning approaches account for less than one-third of the variation in outcomes6163,117. Other markers of injury severity, such as delayed subcortical atrophy, thalamic damage and penetrating injury, have not been measured in PTE studies, but might correlate better with long-term outcomes than do more traditional measures such as neurosurgical interventions, and also correlate closely with PTE76,118,119.

Another limitation of the existing studies is that they rarely assessed whether PTE was well controlled or whether it was medically refractory. Epilepsy that is well controlled with medical therapy is not usually associated with long-term functional decline, whereas medically refractory epilepsy, particularly with temporal lobe onset, has been implicated in progressive neurodegeneration and dementia.

Nevertheless, the temporal relationship between PTE and poor outcomes suggests a causal link. Although early functional outcomes after TBI are similar between patients with and without PTE, the outcomes diverge over time6,111,120. Nearly 15% of patients who have achieved favourable outcomes (GOSE score 5–8) at the time of PTE diagnosis will subsequently decline to unfavourable outcomes121 (GOSE score 2–4). Patients with longer latencies to PTE diagnosis tend to have better outcomes than those with shorter latencies, which might reflect reduced impairment in early recovery121. These temporal trends suggest but do not prove that PTE is an adverse independent prognosticator and second hit after TBI.

PTE is likely to increase long-term mortality risk, although this relationship is similarly confounded by correlations with injury severity122124. Among survivors of the index injury, PTE does not seem to increase mortality during the early post-traumatic period6,122 (1–2 years post-injury). As with functional outcomes, the effects of PTE on mortality seem to be compounded over the long term122124 (years to decades after injury). One study that evaluated death records found that PTE was a leading cause of death in young adults who survived the acute TBI, highlighting a possible causal link between PTE and mortality123.

TBI and epilepsy are notable risk factors for mental health and behavioural problems and cognitive decline. Nearly 60% of survivors of TBI experience psychiatric illness in the year after their injury, and half of patients with epilepsy experience a major depressive episode in their lifetime125,126. PTE and mental health are likely to have a bidirectional relationship: both are associated with damage of emotion-regulating brain networks in the prefrontal and medial brain regions and alter the hypothalamic–pituitary–adrenal axis, thereby affecting mood and behavioural regulation. TBI survivors with PTE are at increased risk of new psychiatric or behavioural conditions, even after accounting for mental health history111. Cognitive networks seem to sustain similar injury patterns, as early seizures and abnormal EEG activity were associated with worse cognitive outcomes in a cohort with similar global outcome measures127,128. In the long term, PTE exacerbates the cognitive decline that is prevalent after TBI114.

Current treatment approaches and outcomes

PTE treatment is highly variable, and the outcome measures are poorly defined, with no formal guidelines for treatment or screening from major societies. Although the onset of PTE mostly occurs during the first year post-TBI, few institutions regularly screen for PTE as part of the routine follow-up. Of specific concern, despite high rates of pharmacoresistance, people with PTE have low rates of referral to specialized epilepsy centres, and surgery is probably underutilized19,108: although it constitutes 20% of epilepsies with structural aetiology, PTE only accounts for 5% of patients in surgical epilepsy clinics72,129.

In clinical practice, PTE is treated similarly to other symptomatic epilepsies, with ASMs as the mainstay of treatment. Literature on the effectiveness of certain ASMs or combinations specifically for PTE is scarce, and owing to historical trials that demonstrated the effectiveness of phenytoin for EPTS prophylaxis, patients with PTE are disproportionately prescribed older-generation ASMs, often by neurotrauma practitioners rather than epilepsy specialists19,20. Newer-generation seizure medications, in particular levetiracetam, might have similar efficacy130 and be better choices given their limited interaction profile and metabolic toxicity and favourable cognitive adverse effect profile131,132. As in other focal epilepsies, pharmacoresistance, defined as the failure of at least two appropriate ASMs to adequately control seizures, represents a major treatment challenge in PTE133. Younger age of PTE onset, focal or mixed onset seizure type, presence of status epilepticus and interictal epileptiform discharges are all predictors of pharmacoresistance80.

Despite the high prevalence of pharmacoresistance, epilepsy surgery evaluations rarely seem to be conducted in PTE populations72,134. This underutilization has been attributed to various factors, including concern about multifocality, distinct pathophysiological mechanisms and anatomical heterogeneity135137. A meta-analysis found higher rates of unfavourable post-surgical outcomes (Engel Epilepsy Surgery Outcome Scale Class II–IV) in temporal lobe PTE than in non-traumatic epilepsy138. Although the result was statistically significant, the absolute reduction in Engel Class I (seizure freedom) rates was 5%, which was not a clinically meaningful difference as the overall Engel Class I rate (70.1%) remained comparable to historical trials139. This result extended to extratemporal lobe PTE, with similarly excellent outcomes reported in patients with focal encephalomalacia on MRI who underwent intracranial EEG evaluations136.

Neuromodulation might also have a role in the treatment of pharmacoresistant PTE. In one study, significant seizure reduction was observed in around two-thirds of patients with this condition who were treated with vagus nerve stimulation140. Similarly, preliminary evidence suggests moderate efficacy of responsive neurostimulation, minimally invasive ablative strategies including laser interstitial therapy, and non-invasive neuromodulation in people with PTE134,141.

Overall, we have a paucity of evidence to guide both medical and surgical care for PTE, with many patients not being referred to specialized care centres.

PTE biomarkers

Although most TBI centres do not screen for PTE, biomarkers hold promise to identify high-risk groups, with direct translational benefit for testing novel therapeutic targets. PTE might be unique among the clinical epilepsies in that it is attributable to a readily identifiable discrete event — that is, the TBI — which initiates epileptogenesis and confers an increased epilepsy risk. This characteristic facilitates the evaluation and identification of biomarkers at specific time points, ranging from hyperacute (minutes) to chronic (years). Two main categories of PTE biomarkers are commonly studied: clinical biomarkers comprising electrophysiological and radiological data, and molecular biomarkers, defined as protein and nucleic acid substrates.

Clinical biomarkers

One of the most intuitive biomarker categories for PTE is EEG. Acute EEG monitoring has become the standard of care for patients with TBI to identify EPTSs and status epilepticus. The presence of epileptiform abnormalities in the acute phase has been linked to increased risk of future development of PTE142,143. Automated detection of these abnormalities further enables broad-scale utilization of these biomarkers144. Findings from human retrospective data sets have been replicated in rodent models, providing additional insights into epileptogenesis145147. In two studies published in 2023, EEG background characteristics, as assessed by quantitative EEG, were also shown to aid PTE prediction in humans142,148.

Structural brain imaging is a crucial component of early TBI evaluation, affording the evaluation of radiological characteristics as biomarkers of PTE. Multiple putative imaging biomarkers, including hippocampal volume, BBB disruption, diffusion tensor imaging and inflammatory and metabolic changes, have been reviewed previously64,65. The Epilepsy Bioinformatics Study for Anti-Epileptogenic Therapy (EpiBioS4Rx) has elucidated several novel imaging biomarkers, including focal cortical and subcortical volume loss118, lesion location and differential volumetrics74 and differential functional connectivity patterns116.

Molecular biomarkers

The unique nature of PTE as a model of epileptogenesis has engendered substantial interest in acute molecular biomarkers, given their potential translatability to other epilepsies. Neuronal, glial and axonal injury, along with inflammatory markers, have all been studied in TBI and previously reviewed149. A systematic review identified blood neuregulin 1 and both blood and cerebrospinal fluid IL-1β as potential PTE biomarkers150. Small-scale genetic association studies have also provided preliminary evidence for a genetic signature of PTE151.

MicroRNAs (miRNAs) — short non-coding RNA sequences that modulate protein synthesis and gene expression at the post-transcriptional level152 — have been identified as tractable targets for biomarker discovery in TBI and PTE. The broad effects of miRNAs on gene and protein expression, coupled with isoform-specific brain selectivity152 and peripheral bioavailability within serum exosomes, has led these RNAs to be implicated in a diverse array of neurological conditions153. miRNAs have been evaluated as diagnostic biomarkers in a range of epilepsies154157, although current data regarding their predictive capacity in PTE are conflicting. The EPITARGET rodent study found that although neuronally enriched plasma miRNAs reflected the extent of cortical injury, they did not predict PTE development158. By contrast, a systematic review evaluated the convergence of miRNA dysregulation in TBI and epilepsy and found 10 specific miRNAs (largely validated in serum) that were common to the two conditions, suggesting roles as PTE biomarkers159.

Exosomes, which provide an important transport mechanism for miRNA, represent another novel class of PTE biomarkers. Exosomes are extracellular vesicles that are rich in bioactive molecules such as proteins and miRNAs, which modulate cellular environmental responses. They can be assayed in peripheral blood and might provide a window into CNS function. Exosome-based biomarkers, predominantly miRNAs, have been evaluated in both TBI160 and epilepsy154,161163. Several groups have shown unique miRNA profiles in humans after TBI164169. Similarly, unique miRNA profiles have been linked to human epilepsy155,170,171. Future work with peripheral exosomes in both preclinical animal models and people with PTE might produce new advances in PTE biomarker discovery.

An important caveat for studies evaluating candidate TBI biomarkers was raised by a preclinical study, which demonstrated that treatments such as prophylactic ASMs given in the acute post-TBI period, as well as EPTSs, can influence plasma protein biomarker levels and, potentially, their predictive value23.

The gut microbiome

The gut microbiome has been explored for its role in the pathogenesis of brain disorders, including epilepsy. A preclinical study in the rat lateral fluid percussion injury model suggested that pre-existing gut microbiome could determine susceptibility to PTE172. A combination of microbiota species of the Lachnospiraceae family and short-chain fatty acids in the faeces predicted PTE with an area under the curve of 0.78 (ref. 172). Importantly, microbiome modification through microbiota transfer had positive effects on PTE risk173.

Summary

PTE biomarkers have a vital role in the elucidation of the mechanisms underlying epileptogenesis and the development of anti-epileptogenic therapeutics. The role of these biomarkers in both diagnosis and measuring treatment response will be invaluable to these pursuits. Given the wide array of clinical and molecular biomarkers, we advocate the use of large-scale, collaborative, multimodal efforts, such as EpiBioS4Rx, for biomarker discovery and validation.

Epileptogenesis in PTE

Mechanisms

Epileptogenesis is the process by which a previously healthy brain tissue develops the propensity to generate spontaneous recurrent seizures. This process is well recognized in both clinical epilepsy and preclinical animal models174,175. PTE provides a model disease condition not only for elucidating the underlying mechanisms but also for potentially halting epileptogenesis. TBI is a discrete inciting event, which is followed by a recognized latency period before the onset of clinical epilepsy (Fig. 2). In humans, the average latency period is several months11 but can extend up to a decade174. As in other epilepsies108, epileptogenesis after TBI does not stop at clinical epilepsy onset; recurrent seizures continually remodel and reinforce seizure networks through strengthening of abnormal circuitry176. This concept is consistent with clinical observations that epilepsy of an individual patient continues to evolve over their disease course, with changes in semiology, seizure frequency and pharmacoresistance.

Fig. 2 |. Mechanisms of epileptogenesis after traumatic brain injury.

Fig. 2 |

Epileptogenesis is the chronic process whereby normally functioning brain tissue is altered to become prone to recurrent abnormal electrical activity that results in seizures. In the case of traumatic brain injury, the initial insult is followed by a latency period, during which a multitude of biological processes result in the formation of epileptogenic circuitry, ultimately leading to post-traumatic epilepsy (PTE). The epileptogenic process continues after seizure onset to reinforce and develop new abnormal circuits, causing the repetitive, spontaneous seizures that characterize PTE. AMPAR, AMPA receptor; NMDAR, NMDA receptor; NOS, nitric oxide synthase; ROS, reactive oxygen species.

A wide range of molecular mechanisms, including neuroinflammation, BBB dysfunction, cell signalling pathway perturbations, astrocyte dysregulation and epigenetic dysregulation, have been implicated in epileptogenesis in PTE. Ultimately, these molecular alterations lead to structural reorganization of the neural circuitry. These structural changes often have significant electrophysiological consequences, especially in PTE originating from the medial temporal lobe. In rodent models, hippocampal hyperexcitability early after TBI results from selective loss of inhibitory interneurons in the dentate gyrus and hilar regions177179. These interneurons help to mediate tonic inhibition through GABA activation of extrasynaptic receptors and are particularly susceptible to loss after TBI180. Loss of these cells, as well as alterations in GABA receptor subunits181,182, changes the inhibitory–excitatory balance, leading to reorganization of circuits183. Reactive plasticity of excitatory dentate granule cells induces aberrant mossy fibre sprouting, connecting dentate granule cell axons with neighbouring neurons and creating recurrent excitatory circuits184,185. In humans, a similar process might occur, with epileptogenesis further reinforcing epileptic circuits, thereby inducing neurodegeneration and gliosis to cause medial temporal PTE. About 30–60% of patients with PTE have seizures originating in the hippocampus8,72.

Inflammation has long been considered as a key driver of epileptogenesis. Seizures and inflammation have a bidirectional relationship, with inflammation inducing seizures and vice versa. Work in animal models has shown that seizures activate nearby glial cells that subsequently release pro-inflammatory cytokines and mediators5,183. As a source of secondary brain damage in the acute phase of TBI, seizures further exacerbate neuronal cell degeneration and initiation of the immune cascade. Within PTE, inflammation is a hallmark of secondary damage after TBI via glial cell activation and migration of inflammatory cells into the CNS5. Microglia are CNS-specialized macrophages, which, when chronically activated, produce cytotoxic molecules that contribute to oxidative stress, and phagocytose normal cells5. Astrocytes, which outnumber neurons at least tenfold in the human brain, maintain neuronal function through buffering of extracellular potassium and recycling of glutamate. Disruption of these functions leads to increased seizure susceptibility and alterations in excitatory circuitry. Astrocytes and microglia are abundant in post-surgical pathological specimens from patients with epilepsy, and they contribute to the recruitment of peripheral leukocytes and lymphocytes that are not typically found in the CNS5,186. Increased activation of CNS-specific and peripheral inflammatory cells might partially explain the vulnerability of younger patients to PTE11,29. Young animals and humans show an increased inflammatory responses to insults, such as altered microglial and cytokine expression patterns, that could increase PTE risk183.

CNS and peripheral inflammatory cells release a plethora of inflammatory molecules that could contribute to PTE. Three signalling pathways, IL-1β, high-mobility group box 1–toll-like receptor 4 (HMGB1–TLR4) and C–C motif chemokine 2 (CCL2), might be drivers of epileptogenesis and are an important focus of research183. IL-1β belongs to the IL-1 family of inflammatory cytokines, which mediate the innate immune response. IL-1β is produced by a range of lymphoid cells and initiates intracellular signalling with mitogen-activated protein kinase, nuclear factor ĸB and other factors to stimulate immune cell activation187. IL-1β is highly expressed both immediately and several months after TBI and correlates with poor outcomes188,189. Antibodies that neutralize this cytokine ameliorate oedema and cognitive deficits in mouse models of TBI190. HMGB1 is a damage-associated molecule that is released by a wide range of CNS cells after tissue necrosis or damage191. HMGB1 was found at high levels in tissues resected during epilepsy surgery192 and probably contributes to epileptogenesis by increasing NMDA receptor function in TLR4-expressing hippocampal neurons193. CCL2, a chemokine receptor that is involved in monocyte recruitment, is produced at high levels by astrocytes within hours of TBI194. It is thought to modulate ion channel expression to induce a pro-excitatory state with decreased neuronal inhibition195. Together, these pathways, along with other inflammatory molecules and cells, produce a pro-inflammatory environment that contributes to epileptogenesis. Although the work on these pathways was conducted in rodent models of epilepsy, surgical sampling of human epileptic tissue seems to confirm the active role of the immune system in epileptogenesis186.

Another epileptogenic mechanism in PTE is BBB dysfunction, which occurs early after TBI, probably as a result of focal structural damage and changes in cerebrovascular permeability196. In other epilepsies, BBB disruption is independently epileptogenic and levels of relevant biomarkers increase at seizure onset197,198. In PTE, BBB disruption induces a pro-seizure state through altered potassium regulation caused by disruption of the potassium gradient, which is maintained by ATP pumps, and spatial buffering, which is maintained by astrocyte foot processes199. The BBB can also be disrupted through alterations in the transforming growth factor β–albumin signalling pathway183. As identified in animal models and verified in human studies, albumin induces a hypersynchronized excitatory response to electrical stimulation200,201, with characteristic EEG alterations and paroxysmal slow cortical activity200. Albumin has been shown to bind to transforming growth factor β receptors on hippocampal neurons, inducing pro-excitatory cytokine production and increasing neuronal excitability183.

Iron accumulation might also contribute to epileptogenesis in PTE. After TBI, intracranial bleeding and cell lysis increase extracellular iron deposition, thereby aggravating injury and inducing epileptogenesis202. Excess iron generates free radicals, which exacerbate damage caused by reactive oxygen species released in the inflammatory cascade. In animal models, iron deposition induces epileptogenesis through upregulation of haem oxygenase 1, which initiates a cascade to release free iron. This process results in paroxysmal EEG discharges and seizure development203. Treatment with iron chelators might alleviate iron-associated damage and reduce PTE risk.

Therapeutic strategies

Alterations in specific signalling pathways after TBI might provide tractable therapeutic targets for PTE (Table 1). The mechanistic target of rapamycin (mTOR) signalling pathway has important roles in neuronal physiology, governing proliferation, growth, synaptic plasticity, morphology and cortical development204,205. mTOR has been best studied in tuberous sclerosis, in which mTOR inhibition reduces seizure and tumour burden. In animal models of PTE, the mTOR inhibitor rapamycin also had a neuroprotective effect, attenuating post-traumatic neurogenesis, hippocampal mossy fibre sprouting and dentate granule cell excitability, but without a reduction in behavioural seizures206. WNT signalling may also have a role in epileptogenesis in PTE. This pathway affects neuronal signalling after TBI207 and was observed to be dysregulated in the hippocampus during epileptogenesis in preclinical animal models208,209.

Table 1 |.

Therapeutic epileptogenic targets in post-traumatic epilepsy

Target Mechanism Potential therapies
Tau hyperphosphorylation Hyperphosphorylation of tau protein to form p-tau is thought to impair neuronal cellular function and contributes to neurodegeneration202 Sodium selenate to reduce p-tau levels22,245,246
Oxidative damage caused by iron toxicity Following brain haemorrhage, iron accumulation from haemolysis leads to generation of free radicals, mitochondrial fragmentation and oxidative damage Iron chelator deferoxamine minocycline247
Neuroinflammation Traumatic brain injury elicits a robust inflammatory response with microglial activation, disruption of the BBB, increased vascular permeability and release of inflammatory cytokines5,248 Anti-inflammatory agents including cyclooxygenase 2 inhibitors or targeted agents such as IL-1R1 antagonists249,250
BBB disruption BBB disruption causes astrocyte dysregulation, as well as release of inflammatory macromolecules and leukocytes, ultimately resulting in progressive synaptic dysfunction Dexamethasone; vascular endothelial growth factor inhibitors251
Epigenetic dysregulation Post-transcriptional dysregulation of gene expression Histone deacetylase inhibitors; DNA methyltransferase inhibitors; engineered site-specific chromatin remodellers252
Calcium channels Disruption of high-voltage and low-voltage calcium channels alters intracellular calcium levels and induces neuronal death through apoptosis, burst firing from pyramidal cells and development of epileptogenic circuits Drugs targeting calcium channels include ethosuximide, which is effective in animal models of absence epilepsy, and ziconotide, an N-type calcium channel blocker
mTOR The mTOR signalling pathway governs neuronal proliferation, growth and morphology, synaptic plasticity and cortical development253,254 mTOR inhibitor rapamycin
WNT WNT signalling is essential for the formation of neuronal circuits and for synaptic plasticity WNT modulators209
Astrocyte dysregulation Astrocytes assist in maintaining neuronal function by buffering extracellular potassium, recycling glutamate for neuronal signalling and regulating the excitatory-inhibitory transmission balance255 Modification of specific gene expression in astrocytes
Gut microbiome The gut-brain axis refers to bidirectional communication between the enteric nervous system and the CNS172,173 Faecal transplantation

BBB, blood–brain barrier; mTOR, mechanistic target of rapamycin.

Modification of ion channels, in particular calcium channels, is another promising therapeutic strategy202. Changes in intracellular calcium levels and calcium homeostasis have been linked to epilepsy in animal and human models210. Several promising AEMs, some of which are currently used in clinical trials, target low-voltage T-type calcium channels that are widely distributed throughout the brain211.

Epigenetic dysregulation at various levels, including chromatin remodelling, histone post-translational modifications, non-coding RNAs and DNA methylation, has also been evaluated as a mechanism of epileptogenesis212, although it has not been studied extensively in PTE. Desbski et al. investigated DNA methylation patterns in three rodent models of epileptogenesis: a lateral fluid-percussion model, focal amygdala stimulation and systemic pilocarpine injection213. They found both common epileptogenic and aetiology-specific DNA methylation signatures. Epigenetic mechanisms are particularly attractive therapeutic targets given the existence of established drugs such as histone deacetylase inhibitors and DNA methyltransferase inhibitors214216. Furthermore, technological advances have created the potential for more targeted epigenetic therapeutics, for example, engineered site-specific chromatin remodellers or CRISPR–Cas9 technology for targeted chromatin modifications217,218.

Given the wide range of targetable mechanisms of epileptogenesis, PTE might serve as a model disease system for AEM development. PTE has a latency period from the initial injury, with epileptogenesis continuing after the first seizure, thereby providing an extended window for treatment. AEMs have the potential to prevent, stop or reverse the development of epilepsy. The ideal AEM would be administered over a molecularly relevant and clinically recognizable treatment window, with durable effects beyond the treatment period and without long-lasting adverse effects219. By contrast, conventional ASM therapy is aimed at reducing seizure burden but not necessarily halting epileptogenesis133. Traditional ASMs have been trialled as anti-epileptogenic agents in humans; however, attempts to repurpose these medications for PTE prevention have largely failed85,95.

No broad-spectrum AEMs are currently clinically available but they are the subject of intensive research in the preclinical arena. Several large multi-institutional consortiums, including the Team Approach to the Prevention and Treatment of PTE (TAPTE), EpiBioS4Rx and EPiXchange, have created strong networks to share ideas and data to develop bench-to-bedside anti-epileptogenic treatments. These groups and others have developed various preclinical models of epileptogenesis, including blunt or focal cortical injury, in a range of animals, from mice to pigs220. These research efforts have led to exciting preclinical proposals for AEMs and other interventions to modify epileptogenesis.

Although no candidate drug for PTE has so far been validated for human use, this condition has far-reaching potential as a model of epileptogenesis. Successful AEM development in the context of human PTE might not only improve global outcomes in TBI but also inform novel therapies that are broadly applicable to all epilepsies.

Conclusions and future priorities

In this Review, we have presented the clinical complexities of PTE and highlighted the role of PTE research in elucidating the mechanisms of epileptogenesis. PTE is common after TBI and is associated with markers of injury severity, including craniotomy and comatose state. Despite its high prevalence and association with adverse neurological outcomes, treatment strategies for PTE are variable and poorly defined. Promising research has identified clinical, EEG, imaging and fluid biomarkers that might aid stratification of PTE risk.

Building on findings from multi-institutional working groups, we propose that PTE is an appropriate model of human epileptogenesis, affording the discovery of both novel biomarkers and treatment strategies, which might be applicable to epilepsy in general. We have highlighted several potential epileptogenic mechanisms in PTE, including neuroinflammation, BBB dysfunction, cell signalling pathway perturbations, astrocyte dysregulation and epigenetic dysregulation. Future prospective, multicentre, collaborative efforts should uncover novel epileptogenic biomarkers, mechanisms and therapies. Successful treatment strategies for PTE will extend the treatment options from symptomatic therapies to disease-modifying anti-epileptogenic therapies, reducing both the development and progression of PTE.

Successful deployment of anti-epileptogenic therapies will rely heavily on appropriate clinical trial design. Classic RCT designs minimize bias and confounding but are expensive and have limited real-world applicability. When considering RCTs for PTE therapeutics, biomarker discovery — both clinical and molecular — is paramount to enable the recruitment of specific high-risk TBI populations, thereby considerably reducing sample sizes. As participants enrolled in this type of study require long-term follow-up for the primary outcome of PTE development, sample size optimization will be invaluable to trial success. Given the established drawbacks of RCTs, other innovative trial designs might need to be considered.

After a long list of failed clinical trial interventions to treat acute TBI and the lack of FDA-approved therapies for TBI, several funders have adapted their paradigms for clinical research funding within the area of acute brain injury and its sequelae. Some have suggested increased use of pragmatic clinical trials221,222 (PCTs) — a strategy that is intended to reflect real-world implementation of a new intervention. In contrast to the explanatory premise of traditional RCTs, PCTs aim to model future therapeutic effectiveness using high-quality real-world evidence223,224. Although attractive in concept, however, PCTs might not address all the needs of the field. In one example of a pragmatic trial design, 387 patients were recruited from real-world clinical practice settings across 47 centres in 18 countries to test the effectiveness of therapeutic hypothermia to reduce intracranial pressure after TBI225. Notably, the nature of the treatment did not allow blinding, and the trial failed to demonstrate an effective functional outcome.

The need for community and stakeholder engagement is a key issue for future PCTs25,226,227. To ensure treatment equity, engaging individuals from socially disadvantaged, underserved and minoritized backgrounds will be vital to the success of prospective studies and translation to trials and future practice228230. Health inequities have been observed at multiple levels among critically ill individuals, including access to care, acute care and post-hospital care outcomes231233. Effective trial design will require community input representing groups that are affected both by PTE itself and by future practice changes that might emerge from the study234236. These approaches have been more extensively explored and implemented in the field of oncology and social medicine, which might help to inform the TBI, epilepsy and critical care research fields237,238.

To successfully develop PTE clinical trials, investigators will need to better understand and define the desired outcomes. Participatory action research (PAR) and community engagement approaches have been used to broaden our understanding of TBI rehabilitation outcomes and long-term comorbidities, to engage with rural communities on their perspectives of neurological outcomes and to evaluate potential participation in long-term outcome studies among individuals living with post-TBI sequelae239241. In addition, some centres used a PAR approach in pharmacological RCTs for post-acute brain injury management242244. True community engagement and PAR should involve patient and community stakeholders from the time of study conceptualization and design to ensure adequate consideration of and input into aspects such as budgeting for further community engagement, institutional review board approval for engagement and perspectives from people and families involved in the study and to devise a plan for public education and knowledge transfer to the affected communities.

We are optimistic for the future of PTE research and treatment. A combination of robust clinical and basic science research initiatives will continue to improve current clinical practice and inform future treatments, and PTE research also holds great promise for anti-epileptogenic therapy discovery and utilization.

Key points.

  • Post-traumatic epilepsy (PTE) is highly prevalent after traumatic brain injury, impairing neurological recovery and leading to worse functional outcomes.

  • Current epilepsy therapeutics symptomatically treat seizures but do not modify epileptogenesis, the process by which brain tissue becomes prone to seizures.

  • The unique nature of PTE, occurring after a well-defined epileptogenic insult, makes it a promising model system for understanding epileptogenesis.

  • Future research in individuals with PTE populations might not only reveal novel mechanisms of epileptogenesis but also enable anti-epileptogenic therapies to be tested.

Acknowledgements

The authors thank Christopher Brown (Indiana University) for his contributions to the design of the figures for this manuscript.

Competing interests

S.M. is the Charles Frost Chair in Neurosurgery and Neurology and partially funded by grants from NIH U54 NS100064 (EpiBioS4Rx), R01-NS43209 and R01-NS127524, the US Department of Defense (W81XWH-22-1-0510, W81XWH-22-1-0210), a pilot grant from the National Institute of Child Health and Human Development (NICHD) centre grant (P50 HD105352) for the Rose F. Kennedy Intellectual and Developmental Disabilities Research Center (RFK-IDDRC), the Heffer Family and the Segal Family Foundations, the Isabelle Rapin and Harold Oaklander Child Neurology Research Fund in the Isabelle Rapin Child Neurology Division and the Abbe Goldstein/Joshua Lurie and Laurie Marsh/Dan Levitz families. He is on the editorial boards of Brain and Development, Paediatric Neurology, Annals of Neurology, MedLink and Physiological Research. He receives compensation from MedLink for his work as Associate Editor; and royalties from books he co-edited. A.G. acknowledges research grant support from NINDS R01-NS127524, US Department of Defense (W81XWH-22-1-0210, W81XWH-22-1-0510, EP220067), a pilot grant from the NICHD centre grant (P50 HD105352) for the RFK-IDDRC, R01-DA019473, R01-AI164864, the Heffer Family and the Segal Family Foundations, the Isabelle Rapin and Harold Oaklander Child Neurology Research Fund in the Isabelle Rapin Child Neurology Division and the Abbe Goldstein/Joshua Lurie and Laurie Marsh/Dan Levitz families. She is the Editor-in-Chief of Epilepsia Open and associate editor of Neurobiology of Disease and receives royalties from Elsevier, Walters Kluwer and MedLink for publications. J.G.-M. receives consulting fees for Zimmer Biomet. D.C. receives compensation as lead editor for the Brain and Life podcast for the American Academy of Neurology and is co-editor of a new textbook on health equity among neurological disorders including chapters on traumatic brain injury and epilepsy. J.F.C. and K.G. accept fees from NeuroOne Medical Technologies Corporation for consulting. The other authors declare no competing interests.

Glossary

Anti-epileptogenic medications

(AEMs). Therapies that ameliorate epileptogenesis, with lasting effects beyond the period of drug exposure.

Anti-seizure medications

(ASMs). Medications that treat seizures but do not modify the process of epileptogenesis or alter the disease course of epilepsy.

Cerebrospinal fluid shunting

A common neurosurgical procedure to drain cerebrospinal fluid, usually from the ventricular system, thereby decreasing intracranial pressure.

Encephalomalacia

A radiological finding denoting an area of brain tissue that has undergone liquefactive necrosis.

Engel Epilepsy Surgery Outcome Scale

A classification scheme of seizure outcomes after epilepsy surgery using four classes: 1, free of disabling seizures; 2, rare disabling seizures; 3, worthwhile improvement; and 4, no worthwhile improvement.

Extended GOS

(GOSE). An extension of the Glasgow Outcome Scale that subdivides the categories of severe disability, moderate disability and good recovery into lower and upper categories.

Glasgow Coma Scale

(GCS). A broadly utilized clinical scale describing the level of consciousness after traumatic injury.

Glasgow Outcome Scale

(GOS). A global scale for functional outcome after brain injury that rates patient status using five categories: dead, vegetative state, severe disability, moderate disability and good recovery.

Mild TBI

Traumatic brain injury with post-impact (may not need resuscitation) Glasgow Coma Scale score 13–15.

Moderate TBI

Traumatic brain injury with post-resuscitation Glasgow Coma Scale score 9–12.

Pathological posturing responses

Stereotypical movements of the trunk and extremities in response to stimuli, typically indicative of significant CNS injury.

Severe TBI

Traumatic brain injury with post-resuscitation Glasgow Coma Scale score ≤ 8.

References

  • 1.Popescu C, Anghelescu A, Daia C & Onose G Actual data on epidemiological evolution and prevention endeavours regarding traumatic brain injury. J. Med Life 8, 272–277 (2015). [PMC free article] [PubMed] [Google Scholar]
  • 2.Frieden TR, Houry D & Baldwin G Report to Congress on traumatic brain injury in the United States: epidemiology and rehabilitation. Centers for Disease Control and Prevention. https://www.cdc.gov/traumaticbraininjury/pdf/TBI_Report_to_Congress_Epi_and_Rehab-a.pdf (2015). [DOI] [PubMed] [Google Scholar]
  • 3.GBD 2016 Traumatic Brain Injury and Spinal Cord Injury Collaborators. Global, regional, and national burden of traumatic brain injury and spinal cord injury, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 18, 56–87 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Farrell JS, Wolff MD & Teskey GC Neurodegeneration and pathology in epilepsy: clinical and basic perspectives. Adv. Neurobiol 15, 317–334 (2017). [DOI] [PubMed] [Google Scholar]
  • 5.Sharma R et al. Neuroinflammation in post-traumatic epilepsy: pathophysiology and tractable therapeutic targets. Brain Sci. 9, 318 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pease M et al. Association of posttraumatic epilepsy with long-term functional outcomes in individuals with severe traumatic brain injury. Neurology 100, e1967–e1975 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Burke J et al. Association of posttraumatic epilepsy with 1-year outcomes after traumatic brain injury. JAMA Netw. Open 4, e2140191 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fordington S & Manford M A review of seizures and epilepsy following traumatic brain injury. J. Neurol 267, 3105–3111 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lucke-Wold BP et al. Traumatic brain injury and epilepsy: underlying mechanisms leading to seizure. Seizure 33, 13–23 (2015). [DOI] [PubMed] [Google Scholar]
  • 10.Annegers JF, Hauser WA, Coan SP & Rocca WA A population-based study of seizures after traumatic brain injuries. N. Engl. J. Med 338, 20–24 (1998). [DOI] [PubMed] [Google Scholar]
  • 11.Pease M et al. Risk factors and incidence of epilepsy after severe traumatic brain injury. Ann. Neurol 92, 663–669 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Schmidt D, Friedman D & Dichter MA Anti-epileptogenic clinical trial designs in epilepsy: issues and options. Neurotherapeutics 11, 401–411 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pitkänen A, Lukasiuk K, Dudek FE & Staley KJ Epileptogenesis. Cold Spring Harb. Perspect. Med 5, a022822 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Löscher W, Potschka H, Sisodiya SM & Vezzani A Drug resistance in epilepsy: clinical impact, potential mechanisms, and new innovative treatment options. Pharmacol. Rev 72, 606–638 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kehne JH, Klein BD, Raeissi S & Sharma S The National Institute of Neurological Disorders and Stroke (NINDS) Epilepsy Therapy Screening Program (ETSP). Neurochem. Res 42, 1894–1903 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.French JA et al. Antiepileptogenesis and disease modification: clinical and regulatory issues. Epilepsia Open 6, 483–492 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Löscher W & Schmidt D Modern antiepileptic drug development has failed to deliver: ways out of the current dilemma. Epilepsia 52, 657–678 (2011). [DOI] [PubMed] [Google Scholar]
  • 18.World Health Organization. Epilepsy: a public health imperative. WHO. https://iris.who.int/bitstream/handle/10665/325440/WHO-MSD-MER-19.2-eng.pdf (2019). [Google Scholar]
  • 19.Gugger JJ et al. Multimodal quality of life assessment in post-9/11 veterans with epilepsy: impact of drug resistance, traumatic brain injury, and comorbidity. Neurology 98, E1761–E1770 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Raymont V et al. Correlates of posttraumatic epilepsy 35 years following combat brain injury. Neurology 75, 224–229 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Vespa PM et al. The epilepsy bioinformatics study for anti-epileptogenic therapy (EpiBioS4Rx) clinical biomarker: study design and protocol. Neurobiol. Dis 123, 110–114 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Saletti PG et al. Tau phosphorylation patterns in the rat cerebral cortex after traumatic brain injury and sodium selenate effects: an Epibios4rx Project 2 study. J. Neurotrauma 41, 222–243 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Saletti PG et al. Early preclinical plasma protein biomarkers of brain trauma are influenced by early seizures and levetiracetam. Epilepsia Open 8, 586–608 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Engel JJ Epileptogenesis, traumatic brain injury, and biomarkers. Neurobiol. Dis 123, 3–7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Correa DJ et al. Applying participatory action research in traumatic brain injury studies to prevent post-traumatic epilepsy. Neurobiol. Dis 123, 137–144 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Duncan D et al. Big data sharing and analysis to advance research in post-traumatic epilepsy. Neurobiol. Dis 123, 127–136 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Christensen J The epidemiology of posttraumatic epilepsy. Semin. Neurol 35, 218–222 (2015). [DOI] [PubMed] [Google Scholar]
  • 28.Thapa A et al. Post-traumatic seizures — a prospective study from a tertiary level trauma center in a developing country. Seizure 19, 211–216 (2010). [DOI] [PubMed] [Google Scholar]
  • 29.Ritter AC et al. Incidence and risk factors of posttraumatic seizures following traumatic brain injury: a Traumatic Brain Injury Model Systems Study. Epilepsia 57, 1968–1977 (2016). [DOI] [PubMed] [Google Scholar]
  • 30.Haltiner AM, Temkin NR & Dikmen SS Risk of seizure recurrence after the first late posttraumatic seizure. Arch. Phys. Med. Rehabil 78, 835–840 (1997). [DOI] [PubMed] [Google Scholar]
  • 31.Ngugi AK, Bottomley C, Kleinschmidt I, Sander JW & Newton CR Estimation of the burden of active and life-time epilepsy: a meta-analytic approach. Epilepsia 51, 883–890 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Thijs RD, Surges R, O’Brien TJ & Sander JW Epilepsy in adults. Lancet 393, 689–701 (2019). [DOI] [PubMed] [Google Scholar]
  • 33.Angeleri F et al. Posttraumatic epilepsy risk factors: one-year prospective study after head injury. Epilepsia 40, 1222–1230 (1999). [DOI] [PubMed] [Google Scholar]
  • 34.Englander J et al. Analyzing risk factors for late posttraumatic seizures: a prospective, multicenter investigation. Arch. Phys. Med. Rehabil 84, 365–373 (2003). [DOI] [PubMed] [Google Scholar]
  • 35.Ferguson PL et al. A population-based study of risk of epilepsy after hospitalization for traumatic brain injury. Epilepsia 51, 891–898 (2010). [DOI] [PubMed] [Google Scholar]
  • 36.Laing J et al. Risk factors and prognosis of early posttraumatic seizures in moderate to severe traumatic brain injury. JAMA Neurol. 79, 334–341 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Christensen J et al. Long-term risk of epilepsy after traumatic brain injury in children and young adults: a population-based cohort study. Lancet 373, 1105–1110 (2009). [DOI] [PubMed] [Google Scholar]
  • 38.Karlander M, Ljungqvist J & Zelano J Post-traumatic epilepsy in adults: a nationwide register-based study. J. Neurol. Neurosurg. Psychiatry 92, 617–621 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.DeGrauw X et al. Epidemiology of traumatic brain injury-associated epilepsy and early use of anti-epilepsy drugs: an analysis of insurance claims data, 2004–2014. Epilepsy Res. 146, 41–49 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Annegers JF et al. Seizures after head trauma: a population study. Neurology 30, 683–689 (1980). [DOI] [PubMed] [Google Scholar]
  • 41.Mahler B et al. Unprovoked seizures after traumatic brain injury: a population-based case–control study. Epilepsia 56, 1438–1444 (2015). [DOI] [PubMed] [Google Scholar]
  • 42.Santos S, Murphy G, Baxter K & Robinson KM Organisational factors affecting the quality of hospital clinical coding. Heal. Inf. Manag 37, 25–37 (2008). [DOI] [PubMed] [Google Scholar]
  • 43.O’Malley KJ et al. Measuring diagnoses: ICD code accuracy. Health Serv. Res 40, 1620–1639 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Langlois JA, Rutland-Brown W & Wald MM The epidemiology and impact of traumatic brain injury: a brief overview. J. Head Trauma Rehabil 21, 375–378 (2006). [DOI] [PubMed] [Google Scholar]
  • 45.Pugh MJV et al. The prevalence of epilepsy and association with traumatic brain injury in veterans of the Afghanistan and Iraq wars. J. Head Trauma Rehabil 30, 29–37 (2015). [DOI] [PubMed] [Google Scholar]
  • 46.Lolk K, Dreier JW & Christensen J Repeated traumatic brain injury and risk of epilepsy: a Danish nationwide cohort study. Brain 144, 875–884 (2021). [DOI] [PubMed] [Google Scholar]
  • 47.Tubi MA et al. Early seizures and temporal lobe trauma predict post-traumatic epilepsy: a longitudinal study. Neurobiol. Dis 123, 115–121 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Salazar AM et al. Epilepsy after penetrating head injury. I. Clinical correlates: a report of the Vietnam Head Injury Study. Neurology 35, 1406–1414 (1985). [DOI] [PubMed] [Google Scholar]
  • 49.The World Bank. World bank country and lending groups. Worldbank.org. https://datahelpdesk.worldbank.org/knowledgebase/articles/906519-world-bank-country-and-lending-groups (2023). [Google Scholar]
  • 50.Ogunrin OA & Adeyekun AA Profile of post-traumatic epilepsy in Benin City, Nigeria. West Afr. J. Med 29, 153–157 (2011). [DOI] [PubMed] [Google Scholar]
  • 51.Rabiu TB & Adetunmbi B Posttraumatic seizures in a rural Nigerian neurosurgical service. World Neurosurg. 104, 367–371 (2017). [DOI] [PubMed] [Google Scholar]
  • 52.Wang XP et al. Development and external validation of a predictive nomogram model of posttraumatic epilepsy: a retrospective analysis. Seizure 88, 36–44 (2021). [DOI] [PubMed] [Google Scholar]
  • 53.Espinosa-Jovel C, Toledano R, Aledo-Serrano Á, García-Morales I & Gil-Nagel A Epidemiological profile of epilepsy in low income populations. Seizure 56, 67–72 (2018). [DOI] [PubMed] [Google Scholar]
  • 54.Gennarelli T, Champion H, Copes W & Sacco W Comparison of mortality, morbidity, and severity of 59,713 head injured patients with 114,447 patients with extracranial injuries. J. Trauma 37, 962–968 (1994). [DOI] [PubMed] [Google Scholar]
  • 55.Eagle SR, Pease M, Nwachuku E, Deng H & Okonkwo DO Prognostic models for traumatic brain injury have good discrimination but poor overall model performance for predicting mortality and unfavorable outcomes. Neurosurgery 92, 137–143 (2023). [DOI] [PubMed] [Google Scholar]
  • 56.Eagle SR et al. Performance of CRASH and IMPACT prognostic models for traumatic brain injury at 12 and 24 months post-injury. Neurotrauma Rep. 4, 118–123 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Siig Hausted Hanna, N. JF & Odgaard L Epilepsy after severe traumatic brain injury: frequency and injury severity. Brain Inj. 34, 889–894 (2020). [DOI] [PubMed] [Google Scholar]
  • 58.Ritter AC et al. Prognostic models for predicting posttraumatic seizures during acute hospitalization, and at 1 and 2 years following traumatic brain injury. Epilepsia 57, 1503–1514 (2016). [DOI] [PubMed] [Google Scholar]
  • 59.Xu T et al. Risk factors for posttraumatic epilepsy: a systematic review and meta-analysis. Epilepsy Behav. 67, 1–6 (2017). [DOI] [PubMed] [Google Scholar]
  • 60.Temkin NR Risk factors for posttraumatic seizures in adults. Epilepsia 44, 18–20 (2003). [DOI] [PubMed] [Google Scholar]
  • 61.Arefan D, Pease M, Eagle SR, Okonkwo DO & Wu S Comparison of machine learning models to predict long-term outcomes after severe traumatic brain injury. Neurosurg. Focus 54, E14 (2023). [DOI] [PubMed] [Google Scholar]
  • 62.Dijkland SA et al. Prognosis in moderate and severe traumatic brain injury: a systematic review of contemporary models and validation studies. J. Neurotrauma 37, 1–13 (2020). [DOI] [PubMed] [Google Scholar]
  • 63.Steyerberg EW et al. Predicting outcome after traumatic brain injury: development and international validation of prognostic scores based on admission characteristics. PLoS Med 5, 1251–1261 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Immonen R et al. Imaging biomarkers of epileptogenecity after traumatic brain injury — preclinical frontiers. Neurobiol. Dis 123, 75–85 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Garner R et al. Imaging biomarkers of posttraumatic epileptogenesis. Epilepsia 60, 2151–2162 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Prince DA et al. Epilepsy following cortical injury: cellular and molecular mechanisms as targets for potential prophylaxis. Epilepsia 50, 30–40 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Jennett WB & Lewin W Traumatic epilepsy after closed head injuries. J. Neurol. Neurosurg. Psychiatry 23, 295–301 (1960). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Al-Haddad SA & Kirollos RA 5-year study of the outcome of surgically treated depressed skull fractures. Ann. R. Coll. Surg. Engl 84, 196–200 (2002). [PMC free article] [PubMed] [Google Scholar]
  • 69.De Reuck J Risk factors for late-onset seizures related to cerebral contusions in adults with a moderate traumatic brain injury. Clin. Neurol. Neurosurg 113, 469–471 (2011). [DOI] [PubMed] [Google Scholar]
  • 70.La Rocca M et al. Distribution and volume analysis of early hemorrhagic contusions by MRI after traumatic brain injury: a preliminary report of the Epilepsy Bioinformatics Study for Antiepileptogenic Therapy (EpiBioS4Rx). Brain Imaging Behav. 15, 2804–2812 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Messori A, Polonara G, Carle F, Gesuita R & Salvolini U Predicting posttraumatic epilepsy with MRI: prospective longitudinal morphologic study in adults. Epilepsia 46, 1472–1481 (2005). [DOI] [PubMed] [Google Scholar]
  • 72.Gupta PK et al. Subtypes of post-traumatic epilepsy: clinical, electrophysiological, and imaging features. J. Neurotrauma 31, 1439–1443 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Won SY et al. A systematic review of epileptic seizures in adults with subdural haematomas. Seizure 45, 28–35 (2017). [DOI] [PubMed] [Google Scholar]
  • 74.La Rocca M et al. Multiplex networks to characterize seizure development in traumatic brain injury patients. Front. Neurosci 14, 591662 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lutkenhoff ES et al. Early brain biomarkers of post-traumatic seizures: initial report of the multicentre epilepsy bioinformatics study for antiepileptogenic therapy (EpiBioS4Rx) prospective study. J. Neurol. Neurosurg. Psychiatry 91, 1154–1157 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Manninen E et al. Acute thalamic damage as a prognostic biomarker for post-traumatic epileptogenesis. Epilepsia 62, 1852–1864 (2021). [DOI] [PubMed] [Google Scholar]
  • 77.Graham NSN, Cole JH, Bourke NJ, Schott JM & Sharp DJ Distinct patterns of neurodegeneration after TBI and in Alzheimer’s disease. Alzheimers Dement. 19, 3065–3077 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Dinkel J et al. Long-term white matter changes after severe traumatic brain injury: a 5-year prospective cohort. Am. J. Neuroradiol 35, 23–29 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zelano J & Westman G Epilepsy after brain infection in adults. Neurology 95, e3213–e3220 (2020). [DOI] [PubMed] [Google Scholar]
  • 80.Yu T, Liu X, Sun L, Wu J & Wang Q Clinical characteristics of post-traumatic epilepsy and the factors affecting the latency of PTE. BMC Neurol. 21, 301 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Candy N, Tsimiklis C, Poonnoose S & Trivedi R The use of antiepileptic medication in early post traumatic seizure prophylaxis at a single institution. J. Clin. Neurosci 69, 198–205 (2019). [DOI] [PubMed] [Google Scholar]
  • 82.DJohn J, Ibrahim R, Patel P, DeHoff K & Kolbe N Administration of levetiracetam in traumatic brain injury: is it warranted? Cureus 12, e9117 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pease M et al. Multicenter and prospective trial of anti-epileptics for early seizure prevention in mild traumatic brain injury with a positive computed tomography scan. Surg. Neurol. Int 13, 241 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lee ST, Lui TN, Wong CW, Yeh YS & Tzaan WC Early seizures after moderate closed head injury. Acta Neurochir. 137, 151–154 (1995). [DOI] [PubMed] [Google Scholar]
  • 85.Temkin NR et al. A randomized, double-blind study of phenytoin for the prevention of post-traumatic seizures. N. Engl. J. Med 323, 497–502 (1990). [DOI] [PubMed] [Google Scholar]
  • 86.Gugger JJ & Diaz-Arrastia R Early posttraumatic seizures — putting things in perspective. JAMA Neurol. 79, 325–326 (2022). [DOI] [PubMed] [Google Scholar]
  • 87.Jennett B Early traumatic epilepsy: incidence and significance after nonmissile injuries. Arch. Neurol 30, 394–398 (1974). [DOI] [PubMed] [Google Scholar]
  • 88.Glaser AC et al. The effect of antiseizure medication administration on mortality and early posttraumatic seizures in critically ill older adults with traumatic brain injury. Neurocrit. Care 37, 538–546 (2022). [DOI] [PubMed] [Google Scholar]
  • 89.Zhao Y, Wu H, Wang X, Li J & Zhang S Clinical epidemiology of posttraumatic epilepsy in a group of Chinese patients. Seizure 21, 322–326 (2012). [DOI] [PubMed] [Google Scholar]
  • 90.Bakr A & Belli A A systematic review of levetiracetam versus phenytoin in the prevention of late post-traumatic seizures and survey of UK neurosurgical prescribing practice of antiepileptic medication in acute traumatic brain injury. Br. J. Neurosurg 32, 237–244 (2018). [DOI] [PubMed] [Google Scholar]
  • 91.Carney N et al. Guidelines for the management of severe traumatic brain injury, fourth edition. Neurosurgery 80, 6–15 (2017). [DOI] [PubMed] [Google Scholar]
  • 92.Wilson CD et al. Early and late posttraumatic epilepsy in the setting of traumatic brain injury: a meta-analysis and review of antiepileptic management. World Neurosurg. 110, e901–e906 (2018). [DOI] [PubMed] [Google Scholar]
  • 93.Sundararajan K, Milne D, Edwards S, Chapman MJ & Shakib S Anti-seizure prophylaxis in critically ill patients with traumatic brain injury in an intensive care unit. Anaesth. Intensive Care 43, 646–651 (2015). [DOI] [PubMed] [Google Scholar]
  • 94.Sun Y et al. Early post-traumatic seizures are associated with valproic acid plasma concentrations and UGT1A6/CYP2C9 genetic polymorphisms in patients with severe traumatic brain injury. Scand. J. Trauma Resusc. Emerg. Med 25, 85 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Temkin NR et al. Valproate therapy for prevention of posttraumatic seizures: a randomized trial. J. Neurosurg 91, 593–600 (1999). [DOI] [PubMed] [Google Scholar]
  • 96.Wang BC et al. Comparative efficacy of prophylactic anticonvulsant drugs following traumatic brain injury: a systematic review and network meta-analysis of randomized controlled trials. PLoS ONE 17, e0265932 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kwon SJ et al. Lacosamide versus phenytoin for the prevention of early post traumatic seizures. J. Crit. Care 50, 50–53 (2019). [DOI] [PubMed] [Google Scholar]
  • 98.Herman ST et al. Consensus statement on continuous EEG in critically ill adults and children, part I: indications. J. Clin. Neurophysiol 32, 87–95 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Vespa P et al. Metabolic crisis occurs with seizures and periodic discharges after brain trauma. Ann. Neurol 79, 579–590 (2016). [DOI] [PubMed] [Google Scholar]
  • 100.Khor D et al. Early seizure prophylaxis in traumatic brain injuries revisited: a prospective observational study. World J. Surg 42, 1727–1732 (2018). [DOI] [PubMed] [Google Scholar]
  • 101.Inglet S et al. Seizure prophylaxis in patients with traumatic brain injury: a single-center study. Cureus 8, 6–8 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hazama A et al. The effect of Keppra prophylaxis on the incidence of early onset, post-traumatic brain injury seizures. Cureus 10, e2674 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pingue V, Mele C & Nardone A Post-traumatic seizures and antiepileptic therapy as predictors of the functional outcome in patients with traumatic brain injury. Sci. Rep 11, 4708 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zaccara G et al. Do antiepileptic drugs increase the risk of infectious diseases? A meta-analysis of placebo-controlled studies. Br. J. Clin. Pharm 83, 1873–1879 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Carpay JA, Aldenkamp AP & van Donselaar CA Complaints associated with the use of antiepileptic drugs: results from a community-based study. Seizure 14, 198–206 (2005). [DOI] [PubMed] [Google Scholar]
  • 106.Lu X & Wang X Hyponatremia induced by antiepileptic drugs in patients with epilepsy. Expert Opin. Drug Saf 16, 77–87 (2017). [DOI] [PubMed] [Google Scholar]
  • 107.Aarabi B, Taghipour M, Haghnegahdar A, Farokhi M & Mobley L Prognostic factors in the occurrence of posttraumatic epilepsy after penetrating head injury suffered during military service. Neurosurg. Focus 8, e1 (2000). [DOI] [PubMed] [Google Scholar]
  • 108.Galanopoulou AS et al. Antiepileptogenesis and disease modification: progress, challenges, and the path forward — Report of the Preclinical Working Group of the 2018 NINDS-sponsored antiepileptogenesis and disease modification workshop. Epilepsia Open 6, 276–296 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Pingue V et al. Impact of seizures and their prophylaxis with antiepileptic drugs on rehabilitation course of patients with traumatic or hemorrhagic brain injury. Front. Neurol 13, 1060008 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Pugh MJ et al. The military injuries: understanding post-traumatic epilepsy study: understanding relationships among lifetime traumatic brain injury history, epilepsy, and quality of life. J. Neurotrauma 38, 2841–2850 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Juengst SB et al. Post-traumatic epilepsy associations with mental health outcomes in the first two years after moderate to severe TBI: a TBI Model Systems analysis. Epilepsy Behav. 73, 240–246 (2017). [DOI] [PubMed] [Google Scholar]
  • 112.Mazzini L et al. Posttraumatic epilepsy: neuroradiologic and neuropsychological assessment of long-term outcome. Epilepsia 44, 569–574 (2003). [DOI] [PubMed] [Google Scholar]
  • 113.Semple BD, Zamani A, Rayner G, Shultz SR & Jones NC Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol. Dis 123, 27–41 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ngadimon IW et al. An interplay between post-traumatic epilepsy and associated cognitive decline: a systematic review. Front. Neurol 13, 827571 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mukherjee S et al. Neuroinflammatory mechanisms of post-traumatic epilepsy. J. Neuroinflammation 17, 193 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.La Rocca M et al. Functional connectivity alterations in traumatic brain injury patients with late seizures. Neurobiol. Dis 179, 106053 (2023). [DOI] [PubMed] [Google Scholar]
  • 117.Pease M et al. Outcome prediction in patients with severe traumatic brain injury using deep learning from head CT scans. Radiology 304, 385–394 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lutkenhoff ES et al. The subcortical basis of outcome and cognitive impairment in TBI: a longitudinal cohort study. Neurology 95, E2398–E2408 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Walker WC et al. Global outcome and late seizures after penetrating versus closed traumatic brain injury: a NIDRR TBI Model Systems study. J. Head Trauma Rehabil 30, 231–240 (2015). [DOI] [PubMed] [Google Scholar]
  • 120.Bushnik T, Englander J, Wright J & Kolakowsky-Hayner SA Traumatic brain injury with and without late posttraumatic seizures: what are the impacts in the post-acute phase: a NIDRR traumatic brain injury model systems study. J. Head Trauma Rehabil 27, 36–44 (2012). [DOI] [PubMed] [Google Scholar]
  • 121.Yu T et al. Predicting global functional outcomes among post-traumatic epilepsy patients after moderate-to-severe traumatic brain injury: development of a prognostic model. Front. Neurol 13, 874491 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Uski J, Lamusuo S, Teperi S, Löyttyniemi E & Tenovuo O Mortality after traumatic brain injury and the effect of posttraumatic epilepsy. Neurology 91, e878–e883 (2018). [DOI] [PubMed] [Google Scholar]
  • 123.Karlander M, Ljungqvist J, Sörbo A & Zelano J Risk and cause of death in post-traumatic epilepsy: a register-based retrospective cohort study. J. Neurol 269, 6014–6020 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Englander J, Bushnik T, Wright JM, Jamison L & Duong TT Mortality in late post-traumatic seizures. J. Neurotrauma 26, 1471–1477 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Rayner G, Jackson GD & Wilson SJ Two distinct symptom-based phenotypes of depression in epilepsy yield specific clinical and etiological insights. Epilepsy Behav. 64, 336–344 (2016). [DOI] [PubMed] [Google Scholar]
  • 126.Ponsford J Anxiety and depression following TBI. in Neurobehavioural Disability and Social Handicap Following Traumatic Brain Injury 2nd edn (eds McMillan TM& Wood RLL) 167–177 (Taylor & Francis, 2017). [Google Scholar]
  • 127.Foreman B et al. Seizures and cognitive outcome after traumatic brain injury: a post hoc analysis. Neurocrit. Care 36, 130–138 (2022). [DOI] [PubMed] [Google Scholar]
  • 128.Lee H et al. Continuous electroencephalography after moderate to severe traumatic brain injury. Crit. Care Med 47, 574–582 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.He X et al. Resective surgery for drug-resistant posttraumatic epilepsy: predictors of seizure outcome. J. Neurosurg 133, 1568–1575 (2019). [DOI] [PubMed] [Google Scholar]
  • 130.Chartrain AG et al. Antiepileptics for post-traumatic seizure prophylaxis after traumatic brain injury. Curr. Pharm. Des 23, 6428–6441 (2017). [DOI] [PubMed] [Google Scholar]
  • 131.Zimmermann LL, Martin RM & Girgis F Treatment options for posttraumatic epilepsy. Curr. Opin. Neurol 30, 580–586 (2017). [DOI] [PubMed] [Google Scholar]
  • 132.Hamed R, Hussein R, Ibraheem S & Jumaily M Comparative study between leviteracetam, phenytoin & carbamazepine in treating post traumatic epilepsy. NeuroQuantology 18, 1–5 (2020). [Google Scholar]
  • 133.Scheffer IE et al. ILAE classification of the epilepsies: position paper of the ILAE Commission for Classification and Terminology. Epilepsia 58, 512–521 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hitti FL et al. Surgical outcomes in post-traumatic epilepsy: a single institutional experience. Oper. Neurosurg 18, 12–18 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Marks DA, Kim J, Spencer DD & Spencer SS Seizure localization and pathology following head injury in patients with uncontrolled epilepsy. Neurology 45, 2051–2057 (1995). [DOI] [PubMed] [Google Scholar]
  • 136.Hakimian S et al. Long-term outcome of extratemporal resection in posttraumatic epilepsy. Neurosurg. Focus 32, E10 (2012). [DOI] [PubMed] [Google Scholar]
  • 137.Schuele SU & Lüders HO Intractable epilepsy: management and therapeutic alternatives. Lancet Neurol. 7, 514–524 (2008). [DOI] [PubMed] [Google Scholar]
  • 138.Ferreira LD, Tabaeizadeh M & Haneef Z Surgical outcomes in post-traumatic temporal lobe epilepsy: a systematic review and meta-analysis. J. Neurotrauma 41, 319–330 (2024). [DOI] [PubMed] [Google Scholar]
  • 139.Wiebe S, Blume WT, Girvin JP & Eliasziw M A randomized, controlled trial of surgery for temporal-lobe epilepsy. N. Engl. J. Med 345, 311–318 (2001). [DOI] [PubMed] [Google Scholar]
  • 140.Lee H-O et al. Effect of vagus nerve stimulation in post-traumatic epilepsy and failed epilepsy surgery: preliminary report. J. Korean Neurosurg. Soc 44, 196–198 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Shen C-C & Jiang J-F Auricular electroacupuncture for late posttraumatic epilepsy after severe brain injury: a retrospective study. Evid. Based Complement Altern. Med 2019, 5798912 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Chen Y et al. Quantitative epileptiform burden and electroencephalography background features predict post-traumatic epilepsy. J. Neurol. Neurosurg. Psychiatry 94, 245–249 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kim JA et al. Epileptiform activity in traumatic brain injury predicts post-traumatic epilepsy. Ann. Neurol 83, 858–862 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Faghihpirayesh R et al. Automatic detection of EEG epileptiform abnormalities in traumatic brain injury using deep learning. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc 2021, 302–305 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Di Sapia R et al. ECoG spiking activity and signal dimension are early predictive measures of epileptogenesis in a translational mouse model of traumatic brain injury. Neurobiol. Dis 185, 106251 (2023). [DOI] [PubMed] [Google Scholar]
  • 146.Kumar U, Li L, Bragin A & Engel JJ Spike and wave discharges and fast ripples during posttraumatic epileptogenesis. Epilepsia 62, 1842–1851 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Li L et al. Spatial and temporal profile of high-frequency oscillations in posttraumatic epileptogenesis. Neurobiol. Dis 161, 105544 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Pease M et al. Predicting post-traumatic epilepsy using admission electroencephalography after severe traumatic brain injury. Epilepsia 64, 1842–1852 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Ghaith HS et al. A literature review of traumatic brain injury biomarkers. Mol. Neurobiol 59, 4141–4158 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Misra S et al. Common pathways of epileptogenesis in patients with epilepsy post-brain injury: findings from a systematic review and meta-analysis. Neurology 101, e2243–e2256 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Cotter D, Kelso A & Neligan A Genetic biomarkers of posttraumatic epilepsy: a systematic review. Seizure 46, 53–58 (2017). [DOI] [PubMed] [Google Scholar]
  • 152.Nam J-W et al. Global analyses of the effect of different cellular contexts on microRNA targeting. Mol. Cell 53, 1031–1043 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Yu Y et al. The role of exosomal microRNAs in central nervous system diseases. Mol. Cell. Biochem 476, 2111–2124 (2021). [DOI] [PubMed] [Google Scholar]
  • 154.Gitaí DLG et al. Extracellular vesicles in the forebrain display reduced miR-346 and miR-331–3p in a rat model of chronic temporal lobe epilepsy. Mol. Neurobiol 57, 1674–1687 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Chen S-D et al. Circulating microRNAs from serum exosomes may serve as a putative biomarker in the diagnosis and treatment of patients with focal cortical dysplasia. Cells 9, 1867 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Brennan GP & Henshall DC microRNAs in the pathophysiology of epilepsy. Neurosci. Lett 667, 47–52 (2018). [DOI] [PubMed] [Google Scholar]
  • 157.Wang Y et al. Circulating microRNAs from plasma small extracellular vesicles as potential diagnostic biomarkers in pediatric epilepsy and drug-resistant epilepsy. Front. Mol. Neurosci 15, 823802 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Heiskanen M et al. Discovery and validation of circulating microRNAs as biomarkers for epileptogenesis after experimental traumatic brain injury — the EPITARGET Cohort. Int. J. Mol. Sci 24, 2823 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Kumar P miRNA dysregulation in traumatic brain injury and epilepsy: a systematic review to identify putative biomarkers for post-traumatic epilepsy. Metab. Brain Dis 38, 749–765 (2023). [DOI] [PubMed] [Google Scholar]
  • 160.Karnati HK et al. Neuronal enriched extracellular vesicle proteins as biomarkers for traumatic brain injury. J. Neurotrauma 36, 975–987 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Lin Z et al. Serum exosomal proteins F9 and TSP-1 as potential diagnostic biomarkers for newly diagnosed epilepsy. Front. Neurosci 14, 737 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Karttunen J, Heiskanen M, Lipponen A, Poulsen D & Pitkänen A Extracellular vesicles as diagnostics and therapeutics for structural epilepsies. Int. J. Mol. Sci 20, 1259 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Upadhya D & Shetty AK Promise of extracellular vesicles for diagnosis and treatment of epilepsy. Epilepsy Behav. 121, 106499 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Redell JB, Moore AN, Ward NH III, Hergenroeder GW & Dash PK Human traumatic brain injury alters plasma microRNA levels. J. Neurotrauma 27, 2147–2156 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Qin X et al. Expression profile of plasma microRNAs and their roles in diagnosis of mild to severe traumatic brain injury. PLoS ONE 13, e0204051 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ko J et al. Diagnosis of traumatic brain injury using miRNA signatures in nanomagnetically isolated brain-derived extracellular vesicles. Lab Chip 18, 3617–3630 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Hicks SD et al. Overlapping microRNA expression in saliva and cerebrospinal fluid accurately identifies pediatric traumatic brain injury. J. Neurotrauma 35, 64–72 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Di Pietro V et al. MicroRNAs as novel biomarkers for the diagnosis and prognosis of mild and severe traumatic brain injury. J. Neurotrauma 34, 1948–1956 (2017). [DOI] [PubMed] [Google Scholar]
  • 169.Bhomia M, Balakathiresan NS, Wang KK, Papa L & Maheshwari RK A panel of serum miRNA biomarkers for the diagnosis of severe to mild traumatic brain injury in humans. Sci. Rep 6, 28148 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Yan S et al. Altered microRNA profiles in plasma exosomes from mesial temporal lobe epilepsy with hippocampal sclerosis. Oncotarget 8, 4136–4146 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Raoof R et al. Dual-center, dual-platform microRNA profiling identifies potential plasma biomarkers of adult temporal lobe epilepsy. eBioMedicine 38, 127–141 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Medel-Matus J-S et al. Susceptibility to epilepsy after traumatic brain injury is associated with preexistent gut microbiome profile. Epilepsia 63, 1835–1848 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Medel-Matus J-S et al. Modification of post-traumatic epilepsy by fecal microbiota transfer. Epilepsy Behav. 134, 108860 (2022). [DOI] [PubMed] [Google Scholar]
  • 174.French JA et al. Characteristics of medial temporal lobe epilepsy: I. Results of history and physical examination. Ann. Neurol 34, 774–780 (1993). [DOI] [PubMed] [Google Scholar]
  • 175.Löscher W The search for new screening models of pharmacoresistant epilepsy: is induction of acute seizures in epileptic rodents a suitable approach? Neurochem. Res 42, 1926–1938 (2017). [DOI] [PubMed] [Google Scholar]
  • 176.Dudek FE & Staley KJ The time course and circuit mechanisms of acquired epileptogenesis. in Jasper’s Basic Mechanisms of the Epilepsies 4th edn (eds Noebels JL et al. ) (Oxford Univ. Press, 2012). [PubMed] [Google Scholar]
  • 177.Santhakumar V, Ratzliff AD, Jeng J, Toth Z & Soltesz I Long-term hyperexcitability in the hippocampus after experimental head trauma. Ann. Neurol 50, 708–717 (2001). [DOI] [PubMed] [Google Scholar]
  • 178.Dudek FE & Spitz M Hypothetical mechanisms for the cellular and neurophysiologic basis of secondary epileptogenesis: proposed role of synaptic reorganization. J. Clin. Neurophysiol 14, 90–101 (1997). [DOI] [PubMed] [Google Scholar]
  • 179.Cantu D et al. Traumatic brain injury increases cortical glutamate network activity by compromising GABAergic control. Cereb. Cortex 25, 2306–2320 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Lowenstein DH, Thomas MJ, Smith DH & McIntosh TK Selective vulnerability of dentate hilar neurons following traumatic brain injury: a potential mechanistic link between head trauma and disorders of the hippocampus. J. Neurosci 12, 4846–4853 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Raible DJ, Frey LC, Cruz Del Angel Y, Russek SJ & Brooks-Kayal AR GABA(A) receptor regulation after experimental traumatic brain injury. J. Neurotrauma 29, 2548–2554 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Gupta A, Elgammal FS, Proddutur A, Shah S & Santhakumar V Decrease in tonic inhibition contributes to increase in dentate semilunar granule cell excitability after brain injury. J. Neurosci 32, 2523–2537 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Webster KM et al. Inflammation in epileptogenesis after traumatic brain injury. J. Neuroinflammation 14, 10 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Hunt RF, Scheff SW & Smith BN Synaptic reorganization of inhibitory hilar interneuron circuitry after traumatic brain injury in mice. J. Neurosci 31, 6880–6890 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Bolkvadze T, Puhakka N & Pitkänen A Epileptogenesis after traumatic brain injury in Plaur-deficient mice. Epilepsy Behav. 60, 187–196 (2016). [DOI] [PubMed] [Google Scholar]
  • 186.Kumar P et al. Single-cell transcriptomics and surface epitope detection in human brain epileptic lesions identifies pro-inflammatory signaling. Nat. Neurosci 25, 956–966 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Srinivasan D, Yen J-H, Joseph DJ & Friedman W Cell type-specific interleukin-1beta signaling in the CNS. J. Neurosci 24, 6482–6488 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Shiozaki T et al. Cerebrospinal fluid concentrations of anti-inflammatory mediators in early-phase severe traumatic brain injury. Shock 23, 406–410 (2005). [DOI] [PubMed] [Google Scholar]
  • 189.Fan L et al. Experimental brain injury induces expression of interleukin-1 beta mRNA in the rat brain. Brain Res. Mol. Brain Res 30, 125–130 (1995). [DOI] [PubMed] [Google Scholar]
  • 190.Clausen F et al. Neutralization of interleukin-1beta modifies the inflammatory response and improves histological and cognitive outcome following traumatic brain injury in mice. Eur. J. Neurosci 30, 385–396 (2009). [DOI] [PubMed] [Google Scholar]
  • 191.Scaffidi P, Misteli T & Bianchi ME Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418, 191–195 (2002). [DOI] [PubMed] [Google Scholar]
  • 192.Shi Y, Zhang L, Teng J & Miao W HMGB1 mediates microglia activation via the TLR4/NF-κB pathway in coriaria lactone induced epilepsy. Mol. Med. Rep 17, 5125–5131 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Chiavegato A, Zurolo E, Losi G, Aronica E & Carmignoto G The inflammatory molecules IL-1β and HMGB1 can rapidly enhance focal seizure generation in a brain slice model of temporal lobe epilepsy. Front. Cell. Neurosci 8, 155 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Semple BD, Kossmann T & Morganti-Kossmann MC Role of chemokines in CNS health and pathology: a focus on the CCL2/CCR2 and CXCL8/CXCR2 networks. J. Cereb. Blood Flow Metab 30, 459–473 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Gosselin RD et al. Constitutive expression of CCR2 chemokine receptor and inhibition by MCP-1/CCL2 of GABA-induced currents in spinal cord neurones. J. Neurochem 95, 1023–1034 (2005). [DOI] [PubMed] [Google Scholar]
  • 196.Patabendige A & Janigro D The role of the blood–brain barrier during neurological disease and infection. Biochem. Soc. Trans 51, 613–626 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Marchi N, Granata T, Alexopoulos A & Janigro D The blood–brain barrier hypothesis in drug resistant epilepsy. Brain 135, e211 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Bargerstock E et al. Is peripheral immunity regulated by blood–brain barrier permeability changes? PLoS ONE 9, e101477 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Dadas A & Janigro D Breakdown of blood–brain barrier as a mechanism of post-traumatic epilepsy. Neurobiol. Dis 123, 20–26 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Weissberg I et al. Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood–brain barrier dysfunction. Neurobiol. Dis 78, 115–125 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Ivens S et al. TGF-beta receptor-mediated albumin uptake into astrocytes is involved in neocortical epileptogenesis. Brain 130, 535–547 (2007). [DOI] [PubMed] [Google Scholar]
  • 202.Saletti PG et al. In search of antiepileptogenic treatments for post-traumatic epilepsy. Neurobiol. Dis 123, 86–99 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Kendirli MT, Rose DT & Bertram EH A model of posttraumatic epilepsy after penetrating brain injuries: effect of lesion size and metal fragments. Epilepsia 55, 1969–1977 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Takei N & Nawa H mTOR signaling and its roles in normal and abnormal brain development. Front. Mol. Neurosci 7, 28 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Lee DY Roles of mTOR signaling in brain development. Exp. Neurobiol 24, 177–185 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Butler CR, Boychuk JA & Smith BN Effects of rapamycin treatment on neurogenesis and synaptic reorganization in the dentate gyrus after controlled cortical impact injury in mice. Front. Syst. Neurosci 9, 163 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Niu L-J, Xu R-X, Zhang P, Du M-X & Jiang X-D Suppression of Frizzled-2-mediated Wnt/Ca2+ signaling significantly attenuates intracellular calcium accumulation in vitro and in a rat model of traumatic brain injury. Neuroscience 213, 19–28 (2012). [DOI] [PubMed] [Google Scholar]
  • 208.Mardones MD & Gupta K Transcriptome profiling of the hippocampal seizure network implicates a role for Wnt signaling during epileptogenesis in a mouse model of temporal lobe epilepsy. Int. J. Mol. Sci 23, 12030 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Gupta K & Schnell E Neuronal network remodeling and Wnt pathway dysregulation in the intra-hippocampal kainate mouse model of temporal lobe epilepsy. PLoS ONE 14, e0215789 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Raza M et al. Evidence that injury-induced changes in hippocampal neuronal calcium dynamics during epileptogenesis cause acquired epilepsy. Proc. Natl Acad. Sci. USA 101, 17522–17527 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Powell KL, Cain SM, Snutch TP & O’Brien TJ Low threshold T-type calcium channels as targets for novel epilepsy treatments. Br. J. Clin. Pharm 77, 729–739 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Conboy K, Henshall DC & Brennan GP Epigenetic principles underlying epileptogenesis and epilepsy syndromes. Neurobiol. Dis 148, 105179 (2021). [DOI] [PubMed] [Google Scholar]
  • 213.Dębski KJ et al. Etiology matters — genomic DNA methylation patterns in three rat models of acquired epilepsy. Sci. Rep 6, 25668 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Nelson ED, Kavalali ET & Monteggia LM Activity-dependent suppression of miniature neurotransmission through the regulation of DNA methylation. J. Neurosci 28, 395–406 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Machnes ZM et al. DNA methylation mediates persistent epileptiform activity in vitro and in vivo. PLoS ONE 8, e76299 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Deutsch SI, Mastropaolo J, Burket JA & Rosse RB An epigenetic intervention interacts with genetic strain differences to modulate the stress-induced reduction of flurazepam’s antiseizure efficacy in the mouse. Eur. Neuropsychopharmacol 19, 398–401 (2009). [DOI] [PubMed] [Google Scholar]
  • 217.Perez-Pinera P et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat. Methods 10, 973–976 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Dominguez AA, Lim WA & Qi LS Beyond editing: repurposing CRISPR-Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell Biol 17, 5–15 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Sasa M A new frontier in epilepsy: novel antiepileptogenic drugs. J. Pharmacol. Sci 100, 487–494 (2006). [DOI] [PubMed] [Google Scholar]
  • 220.Brady RD et al. Modelling traumatic brain injury and posttraumatic epilepsy in rodents. Neurobiol. Dis 123, 8–19 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Ford I & Norrie J Pragmatic trials. N. Engl. J. Med 375, 454–463 (2016). [DOI] [PubMed] [Google Scholar]
  • 222.Casey JD, Beskow LM & Brown J Use of pragmatic and explanatory trial designs in acute care research: lessons from COVID-19. Lancet Respir. Med 10, 700–714 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Braun R It’s been a TRACK-TBI LONG time coming but well worth the wait. Neurology 101, 287–289 (2023). [DOI] [PubMed] [Google Scholar]
  • 224.McCrea MA et al. Functional outcomes over the first year after moderate to severe traumatic brain injury in the prospective, longitudinal TRACK-TBI study. JAMA Neurol. 78, 982–992 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Andrews PJ et al. Therapeutic hypothermia to reduce intracranial pressure after traumatic brain injury: the Eurotherm3235 RCT. Health Technol. Assess 22, 1–134 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Bastian LA et al. Stakeholder engagement in pragmatic clinical trials: emphasizing relationships to improve pain management delivery and outcomes. Pain Med. 21, S13–S20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Morain S & Largent E Think pragmatically: investigators’ obligations to patient-subjects when research is embedded in care. Am. J. Bioeth 23, 10–21 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Diaz V Encouraging participation of minorities in research studies. Ann. Fam. Med 10, 372–373 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Boden-Albala B et al. Use of community-engaged research approaches in clinical interventions for neurologic disorders in the United States. Neurology 101, S27–S46 (2023). [DOI] [PubMed] [Google Scholar]
  • 230.Griffith DM, Towfighi A, Manson SM, Littlejohn EL & Skolarus LE Determinants of inequities in neurologic disease, health, and well-being: the NINDS Social Determinants of Health Framework. Neurology 101, S75–S81 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Danziger J et al. Temporal trends in critical care outcomes in U.S. minority-serving hospitals. Am. J. Respir. Crit. Care Med 201, 681–687 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Kanter GP, Segal AG & Groeneveld PW Income disparities in access to critical care services. Health Aff. 39, 1362–1367 (2020). [DOI] [PubMed] [Google Scholar]
  • 233.Nayfeh A & Fowler RA Understanding patient- and hospital-level factors leading to differences, and disparities, in critical care. Am. J. Respir. Crit. Care Med 201, 642–644 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Brown KE, Fohner AE & Woodahl EL Beyond the individual: community-centric approaches to increase diversity in biomedical research. Clin. Pharmacol. Ther 113, 509–517 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Benizri N, Hallot S, Burns K & Goldfarb M Patient and family representation in randomized clinical trials published in 3 medical and surgical journals: a systematic review. JAMA Netw. Open 5, e2230858 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Gill M et al. Patient and family member-led research in the intensive care unit: a novel approach to patient-centered research. PLoS ONE 11, e0160947 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Fairley R et al. Increasing clinical trial participation of Black women diagnosed with breast cancer. J. Racial Ethn. Health Disparities 10.1007/s40615-023-01644-z (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Barrett NJ et al. Factors associated with biomedical research participation within community-based samples across 3 National Cancer Institute-designated cancer centers. Cancer 126, 1077–1089 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Miles SR et al. Evolution of irritability, anger, and aggression after traumatic brain injury: identifying and predicting subgroups. J. Neurotrauma 38, 1827–1833 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Driver S, Reynolds M & Kramer K Modifying an evidence-based lifestyle programme for individuals with traumatic brain injury. Brain Inj. 31, 1612–1616 (2017). [DOI] [PubMed] [Google Scholar]
  • 241.Sutton KM et al. Engaging individuals with neurological conditions and caregivers in rural communities in a health research team. Prog. Community Health Partnersh 13, 129–139 (2019). [DOI] [PubMed] [Google Scholar]
  • 242.Shimia M et al. A placebo-controlled randomized clinical trial of amantadine hydrochloride for evaluating the functional improvement of patients following severe acute traumatic brain injury. J. Neurosurg. Sci 67, 598–604 (2023). [DOI] [PubMed] [Google Scholar]
  • 243.Morey CE, Cilo M, Berry J & Cusick C The effect of Aricept in persons with persistent memory disorder following traumatic brain injury: a pilot study. Brain Inj. 17, 809–815 (2003). [DOI] [PubMed] [Google Scholar]
  • 244.Jha A et al. A randomized trial of modafinil for the treatment of fatigue and excessive daytime sleepiness in individuals with chronic traumatic brain injury. J. Head Trauma Rehabil 23, 52–63 (2008). [DOI] [PubMed] [Google Scholar]
  • 245.Shultz SR et al. Sodium selenate reduces hyperphosphorylated tau and improves outcomes after traumatic brain injury. Brain 138, 1297–1313 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Liu S-J et al. Sodium selenate retards epileptogenesis in acquired epilepsy models reversing changes in protein phosphatase 2A and hyperphosphorylated tau. Brain 139, 1919–1938 (2016). [DOI] [PubMed] [Google Scholar]
  • 247.Li Z et al. Iron neurotoxicity and protection by deferoxamine in intracerebral hemorrhage. Front. Mol. Neurosci 15, 927334 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Terrone G, Balosso S, Pauletti A, Ravizza T & Vezzani A Inflammation and reactive oxygen species as disease modifiers in epilepsy. Neuropharmacology 167, 107742 (2020). [DOI] [PubMed] [Google Scholar]
  • 249.Serrano GE et al. Ablation of cyclooxygenase-2 in forebrain neurons is neuroprotective and dampens brain inflammation after status epilepticus. J. Neurosci 31, 14850–14860 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Desjardins P et al. Induction of astrocytic cyclooxygenase-2 in epileptic patients with hippocampal sclerosis. Neurochem. Int 42, 299–303 (2003). [DOI] [PubMed] [Google Scholar]
  • 251.Löscher W & Friedman A Structural, molecular, and functional alterations of the blood–brain barrier during epileptogenesis and epilepsy: a cause, consequence, or both? Int. J. Mol. Sci 21, 591 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Henshall DC & Kobow K Epigenetics and epilepsy. Cold Spring Harb. Perspect. Med 5, a022731 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Guo D, Zeng L, Brody DL & Wong M Rapamycin attenuates the development of posttraumatic epilepsy in a mouse model of traumatic brain injury. PLoS ONE 8, e64078 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Galanopoulou AS, Gorter JA & Cepeda C Finding a better drug for epilepsy: the mTOR pathway as an antiepileptogenic target. Epilepsia 53, 1119–1130 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Coulter DA & Steinhäuser C Role of astrocytes in epilepsy. Cold Spring Harb. Perspect. Med 5, a022434 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES