Next Article in Journal
Sleep and Stroke-Related Delirium: A Systematic Review
Previous Article in Journal
To Nap or to Rest? The Influence of a Sixty-Minute Intervention on Verbal and Figural Convergent and Divergent Thinking
Previous Article in Special Issue
The Role of Neurorehabilitation in Post-COVID-19 Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnostic and Therapeutic Approaches in Neurorehabilitation after Traumatic Brain Injury and Disorders of Consciousness

by
Julian Lippert
* and
Adrian G. Guggisberg
Department of Neurology, University Neurorehabilitation, University Hospital Bern, Inselspital, University of Bern, 3012 Bern, Switzerland
*
Author to whom correspondence should be addressed.
Clin. Transl. Neurosci. 2023, 7(3), 21; https://doi.org/10.3390/ctn7030021
Submission received: 28 March 2023 / Revised: 27 July 2023 / Accepted: 4 August 2023 / Published: 9 August 2023
(This article belongs to the Special Issue Neurorehabilitation)

Abstract

:
Severe traumatic brain injury (TBI) may cause disorders of consciousness (DoC) in the form of coma, unresponsive wakefulness syndrome (UWS), or minimally conscious state (MCS). Despite significant advancements made over the last two decades in detecting, predicting, and promoting the recovery of consciousness in TBI patients with DoC, the available diagnostic and treatment choices remain limited. In cases of severe TBI, the dissolution of consciousness both in the acute and post-acute phases constitutes one of the major clinical findings and challenges. In clinical settings, neurologists and neurorehabilitation specialists are called on to discern the level of consciousness in patients who are unable to communicate, and to project outcomes and recommend approaches to treatment. Standards of care are not available to guide clinical decision-making for this population, often leading to inconsistent, inaccurate, and inappropriate care. Recent studies refer to network-based mechanisms of consciousness as a more promising method to predict outcomes and functional recovery. A further goal is the modulation of neural networks underlying awareness and arousal as the main components of consciousness. This review centers on the difficulties in characterizing individuals experiencing post-traumatic DoC and on the recent advancements made in the identification and prognostication of consciousness recovery through the utilization of advanced neuroimaging and electrophysiological techniques as well as biomarkers. Moreover, we discuss new treatment approaches and summarize recent therapeutic recommendations.

1. Introduction

Compared with common neurological disorders worldwide, TBI has the highest incidence and its potential long-term consequences are of major concern for public health [1]. Due to the clinical heterogeneity and complexity of TBI, both initial treatment and rehabilitation constitute a big challenge. Special structural, organizational, and personnel resources of medical facilities are required, as well as close cooperation between participating institutions. The high frequency and extent of impairments in functioning and participation in daily life activities underline the need for neurorehabilitation efforts in TBI patients.
Despite the widespread recognition among clinicians of the importance of suitable post-acute care, only a small proportion of TBI patients receive the rehabilitation services they require [2]. Furthermore, existing evidence indicates that rehabilitation yields greater efficacy when initiated promptly and provided continuously [3]. However, despite cognitive and psychological impairments being reported by approximately one-third of TBI patients, interventions targeting these specific domains were received by only a minority of affected individuals [4].
Longitudinal studies which investigated rehabilitation outcomes spanning up to 30 years after TBI revealed that early and ongoing rehabilitation efforts can solidify the advancements achieved during the acute clinical phase. Furthermore, these interventions demonstrated potential in reducing hospital stays and alleviating the socioeconomic burden associated with TBI [5,6].
Considerable efforts have been made to establish prognostic models capable of providing risk estimates based on a systematic analysis of empirical data, considering various patient and disease characteristics.
Most prognostic models have been developed for patients with moderate to severe TBI. Among the various models, the International Mission on Prognosis and Analysis of Clinical trials in Traumatic brain injury (IMPACT) [7] and Corticoid Randomisation After Significant Head injury (CRASH) [8] are the most widely used prognostic models and are based on the largest cohorts, consisting of 8509 and 10,008 individuals, respectively.
Despite the widespread acceptance of prognostic models in TBI research, their adoption in clinical practice remains limited. Several barriers hinder their clinical implementation, including the extensive clinical investigations required for prognostic estimates and the predominant focus of these models on predictors assessed at presentation. In contrast, clinicians often take into account the patient’s clinical course and progression after admission when making decisions [1].
Currently, the Glasgow Coma Scale Extended (GOSE) is mostly used in depicting functional outcomes, but it falls short of capturing the intricacies of impairments, particularly those stemming from mild TBI. To achieve a more comprehensive assessment, global functional outcome measures should be supplemented with domain-specific instruments. Consequently, various aspects such as persisting post-concussion symptoms, health-related quality of life (HRQOL), depression, and post-traumatic stress disorder (PTSD) have been analyzed in the prognostic context, with a primary focus on patients with mild TBI [9,10].
A recent investigation utilizing data from the CENTER-TBI Core study focused on predicting health-related quality of life (HRQOL) in 2666 adult patients who completed the Quality of Life After Brain Injury-Overall Scale (QOLIBRI) questionnaires at the 6-month mark post-injury. The study revealed that medical and injury-related characteristics played a more significant role in predicting the physical component summary score, whereas patient-related characteristics had greater importance in predicting the mental component summary score and the QOLIBRI scores following TBI [9]. In mild TBI, the prognosis is largely influenced by the individual patient’s pre-injury comorbidities and mental health, rather than in cases of moderate and severe TBI. This observation has been validated in the CENTER-TBI and TRACK-TBI studies, which identified pre-injury health factors (including mental health) and socio-demographic characteristics (such as education, employment, sex, race, ethnicity, and home country) as significant predictors of outcomes in mild TBI cases. However, it is essential to note that injury severity remains one of the most powerful indicators of the Glasgow Outcome Scale Extended (GOSE), indicating that the extent of injury impact on the patient is also relevant in determining global outcomes following mild TBI [11,12,13].
Significant progress has been made in predicting outcomes in TBI through the application of MRI, especially in cases where injuries may not be visible on CT imaging. Both the CENTER-TBI and TRACK-TBI studies reported the presence of structural traumatic abnormalities on MRI in approximately 30% of patients with mild TBI who had a normal CT scan upon presentation [14,15].
The use of advanced MRI techniques, such as diffusion tensor imaging (DTI) and susceptibility-weighted imaging, has proven to be more sensitive in detecting superficial contusions, traumatic axonal injury, and traumatic vascular injury [16,17]. These advancements in MRI technology have significantly improved our ability to identify and characterize TBI-related abnormalities, even when they may not be apparent on traditional CT scans. A recent study demonstrated associations between diffusion abnormalities and various cognitive impairments. Notably, the study revealed significant negative correlations between executive function and structural damage in specific brain regions, such as the corpus callosum, the inferior longitudinal fasciculi, and the middle cerebellar peduncle [18].
Functional MRI (fMRI) [19], MR spectroscopy [19], and electroencephalography (EEG), particularly high-density (HD)-EEG [20], hold the potential to provide supplementary methods for predicting outcomes in TBI. However, these techniques are still in the process of transitioning from research tools to clinically applicable instruments.
Among the technologies to improve the phenotyping of TBI, blood biomarkers have emerged as the most promising candidates for integration into routine clinical care. While previous research primarily concentrated on S100B, recent investigations have explored a group of new brain injury biomarkers (GFAP, NfL, UCH-L1, and tau) in various TBI studies [21].
Taken together, in recent years and due to large observational studies, there has been huge progress in the characterization of TBI, based on advanced neuroimaging and neurophysiological approaches as well as biomarkers [22]. These new diagnostic approaches will facilitate the identification of common disease mechanisms and lead to new therapeutic approaches and improvements in prognosis. However, these advances have not fully entered clinical diagnosis and care. Additional progress is needed in terms of individualized disease management in both acute and post-acute settings.

2. Epidemiology of TBI and Burden of Disease

Former epidemiological studies report a huge variance in the incidence rates of between 979 per 100,000 people in North America and 282.7 per 100,000 people per year in Europe, with considerable variation between member states [23]. Due to methodological variations confounding the comparisons of TBI epidemiology patterns between regions, countries, and continents, the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) has provided estimates of global TBI incidence by using a standardized approach [24]. Recent data from the GBD study estimated the TBI incidence of 369 per 100,000 people (CI 298-401) worldwide (Global Health Data Exchange (https://ghdx.healthdata.org/gbd-results-tool (accessed on 1 August 2022)).
Measures of disease burden as years of lost life (YLLs) and years lived with disability (YLDs) better capture the socioeconomic consequences of TBI, which goes beyond the incidence and fatality rate. The GBD study estimated that TBI was a cause of 8.1 million YLDs in 2016 worldwide, and data from Europe captured from 16 countries were used to estimate that each TBI death is associated with about 24 YLLs [1].
The highest frequency of hospital admissions for TBI is observed in the older population, aged 65 years and above, followed by children and adolescents [25]. This trend is associated with a rising incidence of TBI in older individuals, surpassing population growth rates in certain countries [26]. Compared to younger age groups, older adults are more prone to experiencing TBI resulting from falls, leading to more severe cognitive and functional impairments [27]. Additionally, they may face an increased risk of functional decline during the post-recovery phase [28].
The age group comprising pediatric and adolescent individuals (ranging from 0 to 19 years) demonstrates the second-highest incidence of hospital admissions due to TBI [29]. In the EU, approximately 345 children or adolescents per 100,000 are admitted to hospitals annually, with around 3 per 100,000 succumbing to TBI, leading to approximately 184 YLLs per 100,000 individuals [29]. In the USA, there are an estimated 1 to 2 million cases of mild TBI in children and adolescents each year, and those with a history of concussion have a four-fold higher risk of experiencing a recurrent concussion [30]. Furthermore, children and young people (aged 5 to 18 years) face significantly elevated risks of mental health problems, psychiatric hospitalization, and self-harm following TBI compared to orthopedic injuries [31]. Although the pediatric and adolescent age group exhibits the lowest overall TBI mortality (constituting about 5% of all TBI-related deaths), the burden of these fatalities remains substantial. In the EU, approximately 3000 TBI-related deaths annually result in nearly 200,000 YLLs [32]. These findings underscore the potential for targeted TBI prevention in this population to yield substantial improvements in quality of life and reductions in YLLs. Moreover, the Lancet Neurology Commission on TBI estimates tremendous socioeconomic costs of around US $400 billion annually worldwide. The Commission further states, that TBI remains one of the top three causes of injury-related death and disability up to 2030.
Concerning mortality in moderate and severe TBI, the currently available data are inconsistent, varying from 12% to 44% during acute inpatient care [33]. Moreover, 45% to 87% of mortality in severe TBI is due to a withdrawal of life-sustaining therapy during the acute setting and when DoC continues [34,35,36].

3. Network-Based Mechanisms and Their Dysfunction on Circuitry and Cellular Levels after TBI

TBI mostly induces both localized brain damage and diffuse axonal injury (DAI). Focal injuries arise from direct impact to the head or when the brain collides with the inner surface of the skull, leading to the occurrence of skull fractures or hematomas [37]. Typically, these injuries manifest in specific regions of the brain, including the orbitofrontal, temporal polar, and occipital regions [38].
The pattern of focal brain injuries often demonstrates poor correlation with the clinical impairments observed after TBI [39]. This discrepancy seems to be explained by the presence of DAI which is a prevalent pathological finding across all TBI severities [40,41]. It is nearly ubiquitous in cases of fatal TBI and exhibits a characteristic distribution pattern throughout various brain regions. Particularly severe cases of DAI result in extensive damage, including the corpus callosum and brainstem [42].
Conversely, diffuse multifocal injuries commonly result from rapid acceleration and deceleration forces, causing shear, tensile, and compressive strains. These strains primarily inflict damage on long-distance connections within the white matter through DAI. Additionally, blood vessels can also be affected, leading to diffuse vascular injury [43,44]. Secondary damage can also arise from processes triggered by the initial injury, such as ischemia, elevated intracranial pressure, infection, inflammation, and neurodegeneration [45,46].
Patients with very unfavorable outcomes, such as those in a vegetative state, frequently exhibit damage to the subcortical white matter and/or the thalamic relay nuclei, while cortical damage may be minimal [47]. In contrast, patients with better outcomes show that DAI plays a significant role in determining persistent cognitive impairment, as demonstrated by recent neuroimaging studies [48].
Most studies primarily focusing on the clinical consequences of focal injuries have limited value in predicting clinical outcomes [41]. Questions pertaining to the prediction of clinical outcomes and the recovery of consciousness following severe brain injuries over long-term periods cannot be addressed by examining individual neurons in isolation. To be functionally relevant, information processed at the neuronal level often necessitates integration across various brain regions [49]. Recent research has demonstrated a growing interest in comprehending the organization of brain networks [50]. Emphasis is placed on understanding the disruption of large-scale intrinsic connectivity networks (ICNs), as interactions between these networks are crucial for higher-level cognitive functions such as memory and attention [41]. Furthermore, advancements in neuroimaging techniques have facilitated the examination of these networks in clinical populations [51,52].
ICNs consist of brain regions that exhibit temporally correlated neural activity. The functional architecture of these networks partly reflects their underlying structural connectivity, meaning that regions strongly connected by white matter tracts are likely to possess similar functional properties. This characteristic renders ICNs susceptible to the effects of TBI since DAI often damages long-range white matter tracts that connect nodes within these networks. Consequently, neuroimaging techniques, including fMRI, offer valuable tools for investigating network function in the context of TBI (see neuroimaging and neurophysiological approaches). TBI-induced network dysfunction exhibits intricate patterns and certain underlying principles are becoming evident. These principles can be exemplified by examining the impact of TBI on two well-defined ICNs: the default mode network (DMN) and the salience network (SN) [41].
The DMN comprises brain regions with high metabolic activity and highly coordinated functionality [53]. Its central nodes include the posterior cingulate cortex (PCC) and ventromedial prefrontal cortex (VMPFC). In cases of TBI, various degrees of damage can be observed within the cingulum bundle, a tract encompassing connections between the PCC and VMPFC [54]. This damage is associated with impairments in sustained attention, where patients struggle to maintain focus over time. The extent of damage to the cingulum bundle correlates with worsening attention deficits, which can be predicted by reductions in functional connectivity within the DMN [55]. This observation highlights how damage to ICN connections can disrupt the functional connectivity of the network and impact cognitive functions supported by that network.
Efficient cognitive function relies on coordinated activity between different networks, and abnormal interactions between the SN and DMN are observed following TBI [56]. The SN responds to behaviorally salient external events and appears to signal the need for decreased activity within the DMN [57]. Structural damage to a specific SN tract connecting the right anterior insula to the midline pre-supplementary motor area-dorsal anterior cingulate cortex serves as a robust and specific predictor of the failure to appropriately deactivate the DMN [58].
At the cellular level, compelling evidence indicates that TBI has the potential to initiate neurodegenerative processes, which play a significant role in determining long-term outcomes [59]. TBI can lead to the development of dementias, such as Alzheimer’s disease (AD) and chronic traumatic encephalopathy (CTE), with both single and repetitive injuries predisposing individuals to subsequent cognitive decline [60,61,62]. These findings suggest a prolonged and dynamic element in the pathophysiology of TBI, with interactions among the initial injury, neurodegenerative proteins, and chronic neuroinflammation playing crucial roles [63].
Mechanisms of neurodegeneration following TBI involve the accumulation of amyloid-β plaques and hyperphosphorylated tau neurofibrillary tangles, which are characteristic features of AD [59,63]. Studies in rodents have demonstrated that aggregated tau can initiate the formation of neurofibrillary tangles and spread trans-synaptically from the initially affected brain region to distal but connected areas [64]. The organization of white matter tracts likely constrains the spread of pathogenic proteins, thereby influencing the pattern of neurodegeneration, which may reflect the structure of large-scale ICNs [65].
Neuroinflammation, characterized by the activation of microglia, plays a central role in the neuroinflammatory response to TBI and can persist for months to years after the initial injury [45,66,67]. This persistent neuroinflammation may influence the spread of abnormal proteins and potentially contribute to neurodegeneration following TBI [68]. Neuroinflammatory processes are often observed at the sites of axonal pathology but can also be detected in regions distant from focal injuries as a result of Wallerian degeneration of damaged axons [69].
The relationship between persistent inflammation and the development of neurodegeneration remains unclear, including whether persistent inflammation is a response to pathological features such as amyloid plaques and neurofibrillary tangles or whether unregulated inflammatory responses drive the progression of neurodegeneration [41].

4. Challenges in the Clinical Diagnosis of TBI Patients with DoC

In clinical and research contexts, the Glasgow Coma Scale (GCS) is mostly used to graduate the initial severity of TBI. The simplicity and rapid assessment of the scale led to its international application in the acute treatment setting and is applied both for diagnostic and prognostic applications [70]. The GCS is based on behavioral observations across three subscale scores—eye-opening (score range 1–4), verbal (score range 1–5), and motor (score range 1–6)—that are summed to provide a total score ranging from 3–15 [71]. The total score is intended to reflect the severity of the injury, with scores of 3–8 indicating a severe injury, 9–12 a moderate injury, and 13–15 a mild injury. Moreover, GCS total scores of 3–8 are often used to define coma [72].
However, the GCS total score does not accurately reflect different levels of consciousness and functional outcome at the individual patient level 7.
Previous studies found that GCS subscale scores are more relevant concerning diagnosis [73] and prognosis [74] than the total score. Moreover, different combinations of subscales that sum to the same total score are associated with variable mortality rates [75]. The implication of these findings spans across clinical and research settings where GCS scores are often further collapsed into three broad categories of mild, moderate, and severe TBI. This coarse classification may mischaracterize individual patient prognosis, as evidenced by some ‘‘severe’’ patients having a favorable outcome [76] and some ‘‘mild’’ patients having an unfavorable outcome. In a previously published sub-analysis of the TRACK-TBI study, 53% of 1453 patients classified as mild TBI (GCS score 13–15) reported functional limitations after 12 months [77].
Taken together, global functional outcome measures such as GCS do not precisely characterize the heterogeneous impairments found after TBI. Thus, there is a need for multi-dimensional outcome assessments, which will both better capture the nature of TBI recovery and increase the sensitivity of outcome variables such as residual disability [78].
The recognition that up to 15–20% of patients who appear unresponsive may be covertly conscious [79,80] (i.e., cognitive motor dissociation [81]) has led to a reappraisal of behavioral outcome measures in clinical trials and a search for both clinical assessments for prognostication and new electrophysiologic and imaging biomarkers [82,83].
For the clinical assessment of patients with DoC, standardized neurobehavioral assessments and qualitative bedside examinations should be applied. In an evidence-based review of neurobehavioral rating scales designed specifically for patients with DoC, six scales were identified that seem to be sensitive for detecting conscious awareness [82]. The Coma Recovery Scale-Revised (CRS-R) [84] received the strongest recommendation of those reviewed, on the basis of psychometric properties for clinical assessment [85].
The CRS-R examines all major categories of consciousness in all six domains (Figure 1). These categories are evaluated in a standardized examination protocol. In a clinical setting, the examination should be repeated at regular intervals at least once a week. The individual categories are summed up to a maximum of 23 points.
Prospective studies revealed that an initial score of more than six points on the CRS-R at the beginning of early rehabilitation is a fairly strong predictor of a good outcome [86].
Cognitive motor dissociation represents a relatively new and currently still dynamically changing diagnostic category (Cognitive-Motor-Dissociation, CMD) [81]. Patients with CMD were not able and do not show any intention to communicate despite the most careful clinical examination. So far, there is no generally accepted terminology for this patient group and apart from CMD, the term “non-behavioral MCS” is proposed [87].
However, the combination of detailed clinical examination and assessment with modern techniques such as fMRI, FDG-PET, and HD-EEG supports the diagnostic procedure of a preserved cognitive and processing ability.

5. Neuroimaging and Neurophysiological Approaches to Improve Diagnosis of Different States of Consciousness

While behavioral assessments of DoC such as the CRS-R remain the gold standard at bedside evaluation, neuroimaging permits objective evaluation of structural CNS damage after TBI. Structural imaging provides additional information concerning diagnosis and prognosis, and can serve as surrogate markers for novel therapeutic interventions. While in the acute setting, CT scanning is preferred because of its accessibility, speed of acquisition, and sensitivity to acute hemorrhage of lesion requiring immediate treatment, it is in general limited in detecting neuronal damage compared to standard MRI imaging.
Previous studies have shown the possible prognostic value of ‘classic’ structural MRI sequences to predict DoC outcomes; for example, the presence of corpus callosum and dorsolateral brainstem lesions correlates with a lack of recovery at the group level [88].
Moreover, elaborated techniques such as DTI permit the assessment of structural white matter damage. Studies evaluated the DTI-MRI techniques to predict 1-year functional outcomes in patients with TBI [89] or anoxic [90] injury. Thus, these techniques offer an opportunity to quantify the structural integrity of the white matter on an individual patient level and quantify the primary and secondary axonal damage encountered in DoC [91].

5.1. Electrophysiology

In DoC, a standard 10–20 low-density EEG (LD-EEG) is routinely used for diagnostic purposes in the acute setting. Visual analysis of the LD-EEG can identify epileptiform activity, guide treatment, and help establish prognosis, especially in anoxic cases [92,93]. However, visual analysis of standard LD-EEG showing a global slowing of brain activity fails to differentiate between different stages of DoC, such as UWS or MCS. Therefore, HD-EEG which utilizes a higher spatial sampling of scalp electrodes and a higher spatial resolution is a more accurate diagnostic tool to predict recovery in a clinical setting. Currently, using HD-EEG in a clinical routine is impeded by cost, setup and interpretation time, and lack of specific or sufficient procedural billing codes.
Recent clinical studies investigated the sensitivity and accuracy of quantitative EEG (qEEG) and EEG-based functional connectivity to predict clinical outcomes in DoC patients [94,95]. These studies revealed that patients with reactive EEG signals to external stimuli, larger EEG amplitudes, and stronger activity in the higher-frequency bands (i.e., alpha 7–13 Hz and beta 14–25 Hz) are more likely to have a positive outcome after 3–6 months [96,97]. Elaborating techniques such as the use of machine learning approaches integrating EEG biomarkers, HD-EEG-based functional connectivity and clinical outcome parameters advance the accuracy of outcome prediction and may help to classify DoC patients in terms of rehabilitative potentials and functional recovery.

5.2. FDG-PET

Previous studies investigating brain metabolism in DoC patients used FDG-PET imaging techniques. DoC was mostly associated with hypometabolic areas in the lateral and medial frontoparietal associative cortices [98] and functional recovery was characterized by regained activity in the frontoparietal awareness network [99]. Further studies applied elaborated techniques with automated classifiers to analyze FDG-PET data to evaluate outcome parameters and to distinguish between MCS and UWS [100]. Interestingly, FDG-PET studies could not reliably distinguish between MCS and UWS, but group studies revealed that CRS-R total score correlated with metabolic activity in frontoparietal cortex areas [101].

5.3. Functional MRI

Numerous studies performed fMRI measurements based on spontaneous fluctuations of blood oxygen level-dependent effects (BOLD) during a ‘resting state’ condition and identified various functional networks associated with conscious cognitive activity [102,103]. One of the best-classified networks is the DMN (see network-based mechanisms and their dysfunction after traumatic brain injury) encompassing the posterior and anterior cortical midline structures which are involved in stimulus-independent thought and self-consciousness [104]. The DMN was shown to be absent in brain death [105] but still partially preserved in UWS [106,107], probably reflecting residual structural connectivity [102].
Further fMRI studies investigated the activation of various brain areas after using auditory, tactile, or visual stimuli both in MCS and UWS patients. Though some patients in UWS who exhibited high-level activation in fMRI showed clinical signs of recovery at a long-term follow-up [108,109], the prognostic and diagnostic value of activation patterns in fMRI studies in DoC patients remains controversial. In this respect, further studies which integrate different diagnostic modalities and combine electrophysiological and neuroimaging techniques might help to better understand the neural correlates of consciousness.

5.4. Fluid Biomarkers

Besides neuroimaging parameters, biomarkers have demonstrated their ability to prognosticate late outcomes in TBI patients [110,111]. The CENTER-TBI study identified UCH-L1, S100B, NfL, and total tau as having the greatest incremental value in predicting mortality or unfavorable outcomes [112]. However, it is worth noting that compared to more severe cases of TBI, biomarkers showed reduced predictive capabilities for outcomes in mild TBI cases and for overall incomplete recovery [1].
Biomarkers perform well in predicting CT and MRI positivity compared to clinical decision rules. The combination of GFAP and UCH-L1 has obtained FDA clearance for assisting in the identification of CT-detectable brain lesions within 12 h of mild TBI [113,114]. Moreover, GFAP has shown remarkable predictive capabilities for CT positivity [111,115] and MRI positivity in individuals with normal CT findings [15].
Initial expectations that patterns of biomarker elevation might differentiate focal from diffuse injury [116,117] have not been confirmed, and some data suggest that biomarkers are more correlated with the overall burden of injury rather than with specific pathoanatomical types of TBI [118].
In the context of predicting mortality or unfavorable outcomes, the CENTER-TBI study reported the highest incremental value for UCH-L1, S100B, NfL, and total tau [112]. However, when compared with more severe TBI cases, biomarkers showed reduced predictive abilities for outcomes in mild TBI cases and for overall incomplete recovery [119].

6. Neurorehabilitation of TBI Patients—Established Treatment Options and New Approaches

Despite broad recognition among clinicians of the need for appropriate neurorehabilitation, only a small percentage of TBI patients receive post-acute care. The CENTER-TBI study reported on 1206 TBI patients who suffered from moderate to severe disability 6 months after the incident and approximately 90% of patients had rehabilitation needs. However, only 30% received in-patient rehabilitation, and only 15% received out-patient rehabilitation [6]. Especially in individuals with mild to moderate TBI, the need for post-discharge rehabilitation is often neglected and both TBI patients and their families suffer from a deficiency of both information and post-acute care. In this regard, the CENTER-TBI showed that 90% of centers do not routinely schedule follow-up visits for patients discharged home from the emergency department. Moreover, only 26% of TBI patients received written information or educational material, and only 6% had a follow-up appointment in hospital. Both the CENTER-TBI and TRACK-TBI study revealed that 30% of patients discharged from the emergency department did not attain full recovery after 6 months [120].
Taken together, access to neurorehabilitation and structured follow-up care following TBI remains suboptimal; at the same time, post-acute diagnostic and treatment options for TBI patients, especially those with DoC, are currently limited. Thus, there is a need for increased knowledge about new diagnostic, prognostic, and evidence-based therapeutic approaches and interventions.
Concerning TBI patients with severe DoC, there are neither national nor international evidence-based guidelines for therapeutic interventions available yet. Only in the UK and in the US do current practice recommendations or guidelines exist that include general recommendations for these patients. Those from the American Academy of Neurology (AAN), the American Congress of Rehabilitation Medicine (ACRM), and the National Institute of Disability, Independent Living, and Rehabilitation Research (NIDILRR) include “Practice Guidelines” that emphasize the importance of inpatient multidisciplinary rehabilitation. Moreover, these guidelines emphasize the need for the prevention of secondary complications through early recognition and treatment of medical problems as well as the early detection of available resources. The importance of the diagnosis of the different stages of consciousness applying standardized diagnostic procedures is particularly emphasized, but also the instruction of family members concerning prognosis and evidence-based therapies. The only precise therapeutic recommendation encompasses the usage of amantadin as a pharmacological activation in TBI patients with DoC [121].

6.1. Pharmacologic Therapies

The treatment amantadin was investigated in a placebo-controlled, randomized, double-blind trial in 184 patients 1–4 months after severe TBI [122].
The trial showed a significantly higher rate of behavioral recovery among patients taking amantadine compared to the placebo group.
Besides amantadine, further pharmacologic agents have been used in randomized trials to promote recovery of consciousness in patients with DoC. Dopaminergic drugs have received particular attention because of the widespread expression of dopaminergic receptors in the CNS, including the anterior forebrain mesocircuit, which appears to be essential in the alteration and recovery of consciousness [123,124] (Figure 2).
In patients with prolonged DoC, the administration of dopaminergic agents leads to behavioral and neuroimaging responses, which have been investigated in small studies [125,126]. Nevertheless, large randomized and placebo-controlled studies are still lacking to provide definitive conclusions regarding efficacy.
Interestingly, various studies revealed the stimulating effects of the sedative agent zolpidem in patients with DoC [127,128]. It is assumed that the modulation of GABAA receptors in the globus pallidus interna induces behavioral improvements through inhibition of the mesocircuit [129] (Figure 2). In a double-blinded, placebo-controlled crossover study, 5% out of 84 participants with DoC were identified as definite responders [130]. A further study revealed behavioral improvements in 20% of 84 patients with DoC after administration of zolpidem [131].
Current research focuses on exploring the therapeutic potential of neurotrophins in the management of TBI. Numerous studies have demonstrated that neurotrophins play crucial roles as modulators of neuroinflammation, apoptosis, blood-brain barrier permeability, memory capacity, and neurite regeneration. The expression of neurotrophins following TBI is influenced by factors such as the severity of the injury, genetic polymorphism, and various post-traumatic time points [132].
During the processes of post-traumatic brain remodeling, altered signaling of growth factors, synaptogenesis, angiogenesis, neuronal cell proliferation, gliogenesis, and structural changes in cells contribute to the reorganization and improved functional recovery [133,134,135].
Studies on neuroprotective agents demonstrate their potential to reduce inflammation, free radical production, and cytoskeleton damage, while early production of excitatory amino acids and inflammatory cytokines in the secondary injury cascade disrupts intracellular calcium homeostasis [136].
Interestingly, exogenous administration of neuropeptides, including neurotrophins, into the cerebrospinal fluid (CSF) enhances protective effects and promotes neuronal repair, regeneration, and synaptic sprouting. Several CSF peptides, such as GDNF, PACAP, VEGF, IGF, FGF2, NGF, and BDNF, have exhibited significantly improved neurogenic capacity that is diminished in TBI [137,138,139].
Both human and murine studies have indicated significant upregulation of pro-neurotrophins following brain injuries or degenerative diseases. The binding of these proteins to p75NTR receptors may regulate programmed neuronal cell death in injured or degenerative conditions [140,141].
Additionally, neurotrophins have exhibited potential as sensitive biomarkers for clinical outcomes in patients with TBI [142,143].
Cerebrolysin, a neurotrophin that has been utilized in psychiatry and neurology for over five decades, has shown promise as a therapeutic agent for TBI. It is a non-lipid mixture consisting of free L-amino acids and neuropeptides with low molecular weight, capable of crossing the blood-brain barrier. Through its ability to stimulate natural neuroprotective and neuroregeneration processes, Cerebrolysin can effectively mitigate the short- and long-term consequences of TBI, offering a safe complementary treatment option [144].
In a double-blind trial, a group of 44 individuals between the ages of 19 and 60, with GCS scores ranging from 4 to 11, were assessed for the severity of their injuries using the Clinical Global Impression (CGI). Cognitive function was evaluated using the Syndrom Kurztest (SKT). Over a period of 21 consecutive days, half of the patients received intravenous administration of Cerebrolysin at a dosage of 50 mL, while the other half received a placebo of equal volume intravenously. Assessments were conducted at the start of the study and subsequently on the 7th, 14th, 21st, 42nd, and 63rd days. The clearest improvements in cognitive functions and GCS were observed in patients receiving Cerebrolysin during the second week of treatment [145].
Findings from another study indicate that Cerebrolysin enhances cognitive function, particularly in long-term memory and drawing abilities in patients three months after TBI, as assessed using the Cognitive Abilities Screening Instrument (CASI) [146]. Furthermore, the results of a large retrospective study confirmed the safety profile of Cerebrolysin and demonstrate its beneficial effects in treating patients following brain trauma [147].

6.2. Transcranial Electrical Stimulation

Within the last decade, some smaller clinical trials have studied the ability of transcranial electrical stimulation (tES) to ameliorate symptoms of improved functions in patients with poststroke motor or language deficits, psychiatric disorders, chronic pain [148], or DoC [149]. tES uses weak electrical current (1–2 mA) applied transcranially to modulate cortical excitability. It comprises transcranial direct current stimulation (tDCS) and transcranial alternating current stimulation (tACS). tDCS is thought to increase focal cortical excitability under the stimulating electrodes, whereas tACS entrains neural oscillation to a specific frequency [150,151].
In randomized controlled clinical trials including patients with DoC, tDCS targeted the dorsolateral prefrontal cortex. Only a small percentage of patients demonstrated alternation of consciousness following prefrontal stimulation [151,152,153], and other stimulation targets such as the motor cortex and posterior parietal region yielded smaller effect sizes. To summarize, tES is a safe technique with very few adverse effects reported. The main limitation of its application is currently its moderate and transient clinical effects. Larger randomized controlled trials with dose and duration findings are needed to further evaluate its therapeutic efficacy.

6.3. Transcranial Magnetic Stimulation

Transcranial magnetic stimulation (TMS) consists of an oscillating current, creating a fluctuating magnetic field at the surface of the skull which induces an electric current of the brain cortex. While the efficacy of repetitive TMS (rTMS) has been shown in various disorders including depression, posttraumatic stress disorders, stroke, and neglect [148,154], the effect of rTMS in patients with DoC was not significant [155,156]. These studies on patients with DoC used a 20 Hz stimulation over the motor cortex, but other stimulation sites, including the prefrontal cortex and angular gyrus, have not been tested yet [157]. A more promising approach is the usage of TMS in conjunction with EEG as a diagnostic tool to measure network connectivity [158]. This approach has the potential of a new neurophysiologic biomarker of treatment effects in patients with DoC [159,160].

6.4. Vagus Nerve Stimulation

The approach of stimulating peripheral nerves follows the hypothesis that, through multiple synaptic connections, stimulation of primary sensory neurons can induce neuroplasticity within somatosensory networks and thus modulate network responsiveness [161,162]. Two approaches are followed to promote recovery in patients with DoC: median nerve stimulation (MNS) and vagus nerve stimulation (VNS). Initially, small pilot studies of MNS revealed improvement in the level of consciousness and long-term outcomes [163,164]. These studies were followed by a larger study including 437 patients with severe TBI, which also showed better recovery at 6 months in the group receiving 2 weeks of MNS compared to the control group [165].
VNS is conducted in two different ways: invasive VNS, which is mostly used to treat refractory epilepsy, and noninvasive VNS, which is applied transcutaneously to the auricular branch of the vagus nerve. In both modalities, it is hypothesized that VNS stimulates brainstem, thalamic and cortical activity in a bottom-up manner [166].
Invasive VNS was recently shown to induce recovery of consciousness in a patient with prolonged DoC [167]. For noninvasive VNS, a small case series reported heterogeneous and less clinically apparent treatment effects [168,169]. In analogy to tDCS and TMS treatment approaches, further randomized controlled trials are needed to evaluate the effect of peripheral nerve stimulation on recovery of consciousness.

6.5. Sensory Therapies

Sensory stimulation encompassing tactile and auditory has been administered to patients with DoC for decades [170]. It is postulated that sensory stimulation may enhance neural processes and support neuroplasticity resulting in a re-emergence of consciousness. Music therapy is the most established form of sensory therapy, preferably performed by a music therapist to activate neural networks that mediate attention, emotion, auditory processing, and self-awareness [171,172]. A recently published study that applied fMRI imaging showed that the exposition of different auditory stimulation evoked both in the healthy control group and in the UWS patient group modulation of the auditory network. This study proves that auditory stimulation has the potential to modulate brain activity in patients with DoC [173].
Moreover, a recent meta-analysis gave hints that music therapy may improve functional outcomes in patients with DoC [174].

6.6. Motor Therapy

Growing evidence suggests that early mobilization and namely verticalization using a tilt-table increases vigilance, improves pulmonary gas exchange by reducing atelectasis, and prevents orthostatic hypotension as well as osteoporosis. A small randomized controlled trial revealed that early verticalization during the acute phase improves both short- and long-term functional and neurological outcomes in patients with severe TBI [175].

6.7. Psychoeducational Therapeutic Approaches in Post-TBI Fatigue

Besides DoC, long-lasting physical symptoms including fatigue headaches, impairments of visual perception, and seizures are common and limit the functional recovery among individuals not only with severe but also with mild to moderate TBI. Post-TBI fatigue (PTBIF) is a ubiquitous sequela of TBI, with an estimated prevalence ranging from 21% to 70% [176], irrespective of severity [177].
A recent systematic review describes methylphenidate and melatonin as the only pharmacological agents found to reduce fatigue in randomized controlled trials. Besides pharmacological interventions, walking and water aerobics were effective exercise interventions tested also in randomized controlled studies [178].
Apart from pharmacological and exercise interventions, an interesting new approach based on a mindfulness-based stress reduction (MBSR) program has been described in a randomized, controlled crossover study. Eighteen participants with stroke and eleven patients with TBI were included and treated with MBSR. The study revealed significant improvements were achieved both in mental fatigue as well as in neuropsychological tests [179]. Thus, the study provides evidence that neuroeducation-based approaches might be a promising non-pharmacological treatment option for mental fatigue after TBI.

7. Practice Guidelines in Neurorehabilitation

Numerous systematic reviews summarize evidence-based treatment options for TBI patients with DoC [180,181,182], but these conclusions have not found their way into clinical routine and practice recommendations yet.
The German neurorehabilitation society (DGNR) joined by several national medical societies recently published a guideline (S3 guideline) for the neurological rehabilitation of adults with coma and DoC [183].
The most important and relevant recommendations are based on the guidelines for the diagnosis of coma and other DoC, which were developed by a European expert group commissioned by the European Academy of Neurology (EAN) in 2020 [184].
The expert group decided to adopt only those recommendations that hold high clinical relevance while excluding other recommendations related to more experimental methodologies (e.g., fMRI).
The following main aspects are considered in the guidelines.
Adult patients with severe loss of consciousness due to acute brain injury:
  • should receive multi-professional neurological rehabilitation treatment (level of evidence B);
  • should receive a treatment attempt with amantadine in increasing doses up to 400 mg daily taken orally to improve the state of consciousness (level of evidence B);
  • should be verticalized on a tilt table (level of evidence B);
  • should undergo multisensory stimulation to improve their reactivity to the environment. Especially auditory stimuli with a high degree of emotional and autobiographic reference should be applied in the context of music therapy (level of evidence B);
  • should undergo a therapy attempt with tDCS over the dorsolateral prefrontal cortex (DLPFC) with a duration of 20 min/day over at least 5 days in addition to conventional therapies (level of evidence B).

8. Conclusions

Evidence from recent large observational studies suggests that early and continuous rehabilitation may improve impairments in function and participation in daily life activities and reduce disease burden. Despite broad recognition among clinicians of the need for neurorehabilitation after TBI, structured follow-up care remains still suboptimal not only in low- and middle-income countries but also in high-income countries.
Within the last few years, the pathophysiological understanding of TBI and DoC has been significantly expanded based on elaborate clinical examination models, neuroimaging and electrophysiological methods. A promising approach is the detection of network-based mechanisms for prognosis and outcome assessment as well as for the development and investigation of targeted therapies.
Electrostimulation methods such as TMS, tDCS, and peripheral nerve stimulation are becoming increasingly important as additional therapeutic approaches to established therapies encompassing physical, occupational, speech/language, and neuropsychological therapy.
In the future, new clinical assessments, advanced neuroimaging, and electrophysiological techniques should be addressed more scientifically in order to define new therapeutic approaches and standards.

Author Contributions

Conceptualization, J.L.; writing original draft, J.L.; review and editing, A.G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maas, A.I.R.; Menon, D.K.; Manley, G.T.; Abrams, M.; Åkerlund, C.; Andelic, N.; Aries, M.; Bashford, T.; Bell, M.J.; Bodien, Y.G.; et al. Traumatic brain injury: Progress and challenges in prevention, clinical care, and research. Lancet Neurol. 2022, 21, 1004–1060. [Google Scholar] [CrossRef] [PubMed]
  2. Andelic, N.; Røe, C.; Tenovuo, O.; Azouvi, P.; Dawes, H.; Majdan, M.; Ranta, J.; Howe, E.I.; Wiegers, E.J.; Tverdal, C.; et al. Unmet Rehabilitation Needs after Traumatic Brain Injury across Europe: Results from the CENTER-TBI Study. J. Clin. Med. 2021, 10, 1035. [Google Scholar] [CrossRef] [PubMed]
  3. Andelic, N.; Ye, J.; Tornas, S.; Roe, C.; Lu, J.; Bautz-Holter, E.; Moger, T.; Sigurdardottir, S.; Schanke, A.-K.; Aas, E. Cost-Effectiveness Analysis of an Early-Initiated, Continuous Chain of Rehabilitation after Severe Traumatic Brain Injury. J. Neurotrauma 2014, 31, 1313–1320. [Google Scholar] [CrossRef] [PubMed]
  4. Jacob, L.; Cogné, M.; Tenovuo, O.; Røe, C.; Andelic, N.; Majdan, M.; Ranta, J.; Ylen, P.; Dawes, H.; Azouvi, P.; et al. Predictors of Access to Rehabilitation in the Year Following Traumatic Brain Injury: A European Prospective and Multicenter Study. Neurorehabilit. Neural Repair 2020, 34, 814–830. [Google Scholar] [CrossRef] [PubMed]
  5. Dijkers, M.P.; Marwitz, J.H.; Harrison-Felix, C. Thirty years of national institute on disability, independent living and rehabilitation research traumatic brain injury model system centers research—An update. J. Head Trauma Rehabil. 2018, 33, 363. [Google Scholar] [CrossRef]
  6. Ponsford, J.; Harrison-Felix, C.; Ketchum, J.M.; Spitz, G.; Miller, A.C.; Corrigan, J.D. Outcomes 1 and 2 Years After Moderate to Severe Traumatic Brain Injury: An International Comparative Study. Arch. Phys. Med. Rehabil. 2021, 102, 371–377. [Google Scholar] [CrossRef]
  7. Steyerberg, E.W.; Mushkudiani, N.; Perel, P.; Butcher, I.; Lu, J.; McHugh, G.S.; Murray, G.D.; Marmarou, A.; Roberts, I.; Habbema, J.D.F.; et al. Predicting Outcome after Traumatic Brain Injury: Development and International Validation of Prognostic Scores Based on Admission Characteristics. PLoS Med. 2008, 5, e165. [Google Scholar] [CrossRef] [Green Version]
  8. Collaborators, M.C.T. Predicting outcome after traumatic brain injury: Practical prognostic models based on large cohort of international patients. BMJ 2008, 336, 425–429. [Google Scholar] [CrossRef] [Green Version]
  9. Helmrich, I.R.A.R.; van Klaveren, D.; Dijkland, S.A.; Lingsma, H.F.; Polinder, S.; Wilson, L.; von Steinbuechel, N.; van der Naalt, J.; Maas, A.I.R.; Steyerberg, E.W.; et al. Development of prognostic models for Health-Related Quality of Life following traumatic brain injury. Qual. Life Res. 2021, 31, 451–471. [Google Scholar] [CrossRef]
  10. Stein, M.B.; Jain, S.; Giacino, J.T.; Levin, H.; Dikmen, S.; Nelson, L.D.; Vassar, M.J.; Okonkwo, D.O.; Diaz-Arrastia, R.; Robertson, C.S.; et al. Risk of posttraumatic stress disorder and major depression in civilian patients after mild traumatic brain injury: A TRACK-TBI study. JAMA Psychiatry 2019, 76, 249–258. [Google Scholar] [CrossRef]
  11. Jacobs, B.; Beems, T.; Stulemeijer, M.; van Vugt, A.B.; van der Vliet, T.M.; Borm, G.F.; Vos, P.E.; Pérez, D.A.; Gallardo, A.J.L.; Agramonte, J.A.D.; et al. Outcome Prediction in Mild Traumatic Brain Injury: Age and Clinical Variables Are Stronger Predictors than CT Abnormalities. J. Neurotrauma 2010, 27, 655–668. [Google Scholar] [CrossRef] [Green Version]
  12. Lingsma, H.F.; Yue, J.K.; Maas, A.I.; Steyerberg, E.W.; Manley, G.T.; Cooper, S.R.; Dams-O’Connor, K.; Gordon, W.A.; Menon, D.K.; Mukherjee, P.; et al. Outcome Prediction after Mild and Complicated Mild Traumatic Brain Injury: External Validation of Existing Models and Identification of New Predictors Using the TRACK-TBI Pilot Study. J. Neurotrauma 2015, 32, 83–94. [Google Scholar] [CrossRef]
  13. Howe, E.I.; Zeldovich, M.; Andelic, N.; von Steinbuechel, N.; Fure, S.C.R.; Borgen, I.M.H.; Forslund, M.V.; Hellstrøm, T.; Søberg, H.L.; Sveen, U.; et al. Rehabilitation and outcomes after complicated vs uncomplicated mild TBI: Results from the CENTER-TBI study. BMC Health Serv. Res. 2022, 22, 1536. [Google Scholar] [CrossRef]
  14. Steyerberg, E.W.; Wiegers, E.; Sewalt, C.; Buki, A.; Citerio, G.; De Keyser, V.; Ercole, A.; Kunzmann, K.; Lanyon, L.; Lecky, F.; et al. Case-mix, care pathways, and outcomes in patients with traumatic brain injury in CENTER-TBI: A European prospective, multicentre, longitudinal, cohort study. Lancet Neurol. 2019, 18, 923–934. [Google Scholar] [CrossRef]
  15. Yue, J.K.; Yuh, E.L.; Korley, F.K.; Winkler, E.A.; Sun, X.; Puffer, R.C.; Deng, H.; Choy, W.; Chandra, A.; Taylor, S.R.; et al. Association between plasma GFAP concentrations and MRI abnormalities in patients with CT-negative traumatic brain injury in the TRACK-TBI cohort: A prospective multicentre study. Lancet Neurol. 2019, 18, 953–961. [Google Scholar] [CrossRef]
  16. Amyot, F.; Arciniegas, D.B.; Brazaitis, M.P.; Curley, K.C.; Diaz-Arrastia, R.; Gandjbakhche, A.; Herscovitch, P.; Hinds, S.R., 2rd; Manley, G.T.; Pacifico, A.; et al. A Review of the Effectiveness of Neuroimaging Modalities for the Detection of Traumatic Brain Injury. J. Neurotrauma 2015, 32, 1693–1721. [Google Scholar] [CrossRef]
  17. Sandsmark, D.K.; Bashir, A.; Wellington, C.L.; Diaz-Arrastia, R. Cerebral Microvascular Injury: A Potentially Treatable Endophenotype of Traumatic Brain Injury-Induced Neurodegeneration. Neuron 2019, 103, 367–379. [Google Scholar] [CrossRef]
  18. Jolly, A.E.; Bălăeţ, M.; Azor, A.; Friedland, D.; Sandrone, S.; Graham, N.S.N.; Zimmerman, K.; Sharp, D.J. Detecting axonal injury in individual patients after traumatic brain injury. Brain 2021, 144, 92–113. [Google Scholar] [CrossRef]
  19. Palacios, E.M.; Yuh, E.L.; Chang, Y.-S.; Yue, J.K.; Schnyer, D.M.; Okonkwo, D.O.; Valadka, A.B.; Gordon, W.A.; Maas, A.I.R.; Vassar, M.; et al. Resting-State Functional Connectivity Alterations Associated with Six-Month Outcomes in Mild Traumatic Brain Injury. J. Neurotrauma 2017, 34, 1546–1557. [Google Scholar] [CrossRef] [Green Version]
  20. O’donnell, A.; Pauli, R.; Banellis, L.; Sokoliuk, R.; Hayton, T.; Sturman, S.; Veenith, T.; Yakoub, K.M.; Belli, A.; Chennu, S.; et al. The prognostic value of resting-state EEG in acute post-traumatic unresponsive states. Brain Commun. 2021, 3, fcab017. [Google Scholar] [CrossRef]
  21. Mondello, S.; Sorinola, A.; Czeiter, E.; Vámos, Z.; Amrein, K.; Synnot, A.; Donoghue, E.; Sándor, J.; Wang, K.; Diaz-Arrastia, R.; et al. Blood-Based Protein Biomarkers for the Management of Traumatic Brain Injuries in Adults Presenting to Emergency Departments with Mild Brain Injury: A Living Systematic Review and Meta-Analysis. J. Neurotrauma 2021, 38, 1086–1106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Navarro, J.C.; Mejia, L.L.P.; Robertson, C. A Precision Medicine Agenda in Traumatic Brain Injury. Front. Pharmacol. 2022, 13, 713100. [Google Scholar] [CrossRef] [PubMed]
  23. Maas, A.I.R.; Menon, D.K.; Adelson, P.D.; Andelic, N.; Bell, M.J.; Belli, A.; Bragge, P.; Brazinova, A.; Büki, A.; Chesnut, R.M.; et al. Traumatic brain injury: Integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017, 16, 987–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. James, S.L.; Theadom, A.; Ellenbogen, R.G.; Bannick, M.S.; Montjoy-Venning, W.; Lucchesi, L.R.; Abbasi, N.; Abdulkader, R.; Abraha, H.N.; Adsuar, J.C.; et al. 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. 2019, 18, 56–87. [Google Scholar] [CrossRef] [Green Version]
  25. Majdan, M.; Plancikova, D.; Brazinova, A.; Rusnak, M.; Nieboer, D.; Feigin, V.L.; Maas, A. Epidemiology of traumatic brain injuries in Europe: A cross-sectional analysis. Lancet Public Health 2016, 1, e76–e83. [Google Scholar] [CrossRef]
  26. Dams-O’Connor, K.; Cuthbert, J.P.; Whyte, J.; Corrigan, J.D.; Faul, M.; Harrison-Felix, C. Traumatic Brain Injury among Older Adults at Level I and II Trauma Centers. J. Neurotrauma 2013, 30, 2001–2013. [Google Scholar] [CrossRef] [Green Version]
  27. Gardner, R.C.; Dams-O’Connor, K.; Morrissey, M.R.; Manley, G.T. Geriatric Traumatic Brain Injury: Epidemiology, Outcomes, Knowledge Gaps, and Future Directions. J. Neurotrauma 2018, 35, 889–906. [Google Scholar] [CrossRef]
  28. Dams-O’Connor, K.; Ketchum, J.M.; Cuthbert, J.P.; Corrigan, J.D.; Hammond, F.M.; Haarbauer-Krupa, J.; Kowalski, R.G.; Miller, A.C. Functional Outcome Trajectories Following Inpatient Rehabilitation for TBI in the United States: A NIDILRR TBIMS and CDC Interagency Collaboration. J. Head Trauma Rehabil. 2020, 35, 127. [Google Scholar] [CrossRef]
  29. Majdan, M.; Melichova, J.; Plancikova, D.; Sivco, P.; Maas, A.I.R.; Feigin, V.L.; Polinder, S.; Haagsma, J.A. Burden of Traumatic Brain Injuries in Children and Adolescents in Europe: Hospital Discharges, Deaths and Years of Life Lost. Children 2022, 9, 105. [Google Scholar] [CrossRef]
  30. van Ierssel, J.; Osmond, M.; Hamid, J.; Sampson, M.; Zemek, R. What is the risk of recurrent concussion in children and adolescents aged 5–18 years? A systematic review and meta-analysis. Br. J. Sports Med. 2021, 55, 663–669. [Google Scholar] [CrossRef]
  31. Ledoux, A.-A.; Webster, R.J.; Clarke, A.E.; Fell, D.B.; Knight, B.D.; Gardner, W.; Cloutier, P.; Gray, C.; Tuna, M.; Zemek, R. Risk of Mental Health Problems in Children and Youths Following Concussion. JAMA Netw. Open 2022, 5, e221235. [Google Scholar] [CrossRef]
  32. Majdan, M.; Plancikova, D.; Maas, A.; Polinder, S.; Feigin, V.; Theadom, A.; Rusnak, M.; Brazinova, A.; Haagsma, J. Years of life lost due to traumatic brain injury in Europe: A cross-sectional analysis of 16 countries. PLoS Med. 2017, 14, e1002331. [Google Scholar] [CrossRef] [Green Version]
  33. Jochems, D.; van Wessem, K.J.P.; Houwert, R.M.; Brouwers, H.B.; Dankbaar, J.W.; van Es, M.A.; Geurts, M.; Slooter, A.J.C.; Leenen, L.P.H. Outcome in Patients with Isolated Moderate to Severe Traumatic Brain Injury. Crit. Care Res. Pract. 2018, 2018, 3769418. [Google Scholar] [CrossRef] [Green Version]
  34. Turgeon, A.F.; Lauzier, F.; Simard, J.-F.; Scales, D.C.; Burns, K.E.; Moore, L.; Zygun, D.A.; Bernard, F.; Meade, M.O.; Dung, T.C.; et al. Mortality associated with withdrawal of life-sustaining therapy for patients with severe traumatic brain injury: A Canadian multicentre cohort study. Can. Med. Assoc. J. 2011, 183, 1581–1588. [Google Scholar] [CrossRef] [Green Version]
  35. Robertsen, A.; Førde, R.; Skaga, N.O.; Helseth, E. Treatment-limiting decisions in patients with severe traumatic brain injury in a Norwegian regional trauma center. Scand. J. Trauma Resusc. Emerg. Med. 2017, 25, 44. [Google Scholar] [CrossRef] [Green Version]
  36. Verkade, M.A.; Epker, J.L.; Nieuwenhoff, M.D.; Bakker, J.; Kompanje, E.J.O. Withdrawal of Life-Sustaining Treatment in a Mixed Intensive Care Unit: Most Common in Patients with Catastropic Brain Injury. Neurocritical Care 2012, 16, 130–135. [Google Scholar] [CrossRef]
  37. Graham, D.I.; McIntosh, T.K.; Maxwell, W.L.; Nicoll, J.A.R. Recent advances in neurotrauma. J. Neuropathol. Exp. Neurol. 2000, 59, 641–651. [Google Scholar] [CrossRef] [Green Version]
  38. Gurdjian, E.S. Re-evaluation of the biomechanics of blunt impact injury of the head. Surg. Gynecol. Obstet. 1975, 140, 845–850. [Google Scholar]
  39. Bigler, E.D. Anterior and middle cranial fossa in traumatic brain injury: Relevant neuroanatomy and neuropathology in the study of neuropsychological outcome. Neuropsychology 2007, 21, 515–531. [Google Scholar] [CrossRef] [Green Version]
  40. Blumbergs, P.; Scott, G.; Manavis, J.; Wainwright, H.; Simpson, D.; McLean, A. Stalning af amyloid percursor protein to study axonal damage in mild head Injury. Lancet 1994, 344, 1055–1056. [Google Scholar] [CrossRef]
  41. Sharp, D.J.; Scott, G.; Leech, R. Network dysfunction after traumatic brain injury. Nat. Rev. Neurol. 2014, 10, 156–166. [Google Scholar] [CrossRef] [PubMed]
  42. Adams, J.H.; Doyle, D.; Ford, I.; Gennarelli, T.A.; Graham, D.I.; Mclellan, D.R. Diffuse axonal injury in head injury: Definition, diagnosis and grading. Histopathology 1989, 15, 49–59. [Google Scholar] [CrossRef] [PubMed]
  43. Smith, D.H.; Meaney, D.F.; Shull, W.H. Diffuse axonal injury in head trauma. J. Head Trauma Rehabil. 2003, 18, 307–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Werner, C.; Engelhard, K. Pathophysiology of traumatic brain injury. Br. J. Anaesth. 2007, 99, 4–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ramlackhansingh, A.F.; Brooks, D.J.; Greenwood, R.J.; Bose, S.K.; Turkheimer, F.E.; Kinnunen, K.M.; Gentleman, S.; Heckemann, R.A.; Gunanayagam, K.; Gelosa, G.; et al. Inflammation after trauma: Microglial activation and traumatic brain injury. Ann. Neurol. 2011, 70, 374–383. [Google Scholar] [CrossRef]
  46. Johnson, V.E.; Stewart, W.; Smith, D.H. Axonal pathology in traumatic brain injury. Exp. Neurol. 2013, 246, 35–43. [Google Scholar] [CrossRef] [Green Version]
  47. Adams, J.H.; Graham, D.I.; Jennett, B. The neuropathology of the vegetative state after an acute brain insult. Brain 2000, 123, 1327–1338. [Google Scholar] [CrossRef] [Green Version]
  48. Kinnunen, K.M.; Greenwood, R.; Powell, J.H.; Leech, R.; Hawkins, P.C.; Bonnelle, V.; Patel, M.C.; Counsell, S.J.; Sharp, D.J. White matter damage and cognitive impairment after traumatic brain injury. Brain 2011, 134, 449–463. [Google Scholar] [CrossRef]
  49. Mesulam, M.-M. From sensation to cognition. Brain J. Neurol. 1998, 121, 1013–1052. [Google Scholar] [CrossRef]
  50. Bullmore, E.; Sporns, O. Complex brain networks: Graph theoretical analysis of structural and functional systems. Nat. Rev. Neurosci. 2009, 10, 186–198. [Google Scholar] [CrossRef]
  51. Beckmann, C.F.; DeLuca, M.; Devlin, J.T.; Smith, S.M. Investigations into resting-state connectivity using independent component analysis. Philos. Trans. R. Soc. B Biol. Sci. 2005, 360, 1001–1013. [Google Scholar] [CrossRef] [Green Version]
  52. Smith, S.M.; Fox, P.T.; Miller, K.L.; Glahn, D.C.; Fox, P.M.; Mackay, C.E.; Filippini, N.; Watkins, K.E.; Toro, R.; Laird, A.R.; et al. Correspondence of the brain’s functional architecture during activation and rest. Proc. Natl. Acad. Sci. USA 2009, 106, 13040–13045. [Google Scholar] [CrossRef]
  53. Raichle, M.E.; MacLeod, A.M.; Snyder, A.Z.; Powers, W.J.; Gusnard, D.A.; Shulman, G.L. A default mode of brain function. Proc. Natl. Acad. Sci. USA 2001, 98, 676–682. [Google Scholar] [CrossRef]
  54. Greicius, M.D.; Supekar, K.; Menon, V.; Dougherty, R.F. Resting-State Functional Connectivity Reflects Structural Connectivity in the Default Mode Network. Cereb. Cortex 2008, 19, 72–78. [Google Scholar] [CrossRef]
  55. Bonnelle, V.; Leech, R.; Kinnunen, K.M.; Ham, T.E.; Beckmann, C.F.; De Boissezon, X.; Greenwood, R.J.; Sharp, D.J. Default Mode Network Connectivity Predicts Sustained Attention Deficits after Traumatic Brain Injury. J. Neurosci. 2011, 31, 13442–13451. [Google Scholar] [CrossRef] [Green Version]
  56. Bonnelle, V.; Ham, T.E.; Leech, R.; Kinnunen, K.M.; Mehta, M.A.; Greenwood, R.J.; Sharp, D.J. Salience network integrity predicts default mode network function after traumatic brain injury. Proc. Natl. Acad. Sci. USA 2012, 109, 4690–4695. [Google Scholar] [CrossRef]
  57. Seeley, W.W.; Menon, V.; Schatzberg, A.F.; Keller, J.; Glover, G.H.; Kenna, H.; Reiss, A.L.; Greicius, M.D. Dissociable Intrinsic Connectivity Networks for Salience Processing and Executive Control. J. Neurosci. 2007, 27, 2349–2356. [Google Scholar] [CrossRef] [Green Version]
  58. Sharp, D.; Bonnelle, V.; De Boissezon, X.; Beckmann, C.F.; James, S.G.; Patel, M.C.; Mehta, M.A. Distinct frontal systems for response inhibition, attentional capture, and error processing. Proc. Natl. Acad. Sci. USA 2010, 107, 6106–6111. [Google Scholar] [CrossRef]
  59. Smith, D.H.; Johnson, V.E.; Stewart, W. Chronic neuropathologies of single and repetitive TBI: Substrates of dementia? Nat. Rev. Neurol. 2013, 9, 211–221. [Google Scholar] [CrossRef] [Green Version]
  60. Lye, T.C.; Shores, E.A. Traumatic brain injury as a risk factor for Alzheimer’s disease: A review. Neuropsychol. Rev. 2000, 10, 115–129. [Google Scholar] [CrossRef]
  61. Goldstein, L.E.; Fisher, A.M.; Tagge, C.A.; Zhang, X.L.; Velisek, L.; Sullivan, J.A.; Upreti, C.; Kracht, J.M.; Ericsson, M.; Wojnarowicz, M.W.; et al. Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci. Transl. Med. 2012, 4, ra134–ra160. [Google Scholar]
  62. Sayed, N.; Culver, C.; Dams-O’Connor, K.; Hammond, F.; Diaz-Arrastia, R.; Guetta, G.; Hahn-Ketter, A.E.; Fedor, A.; Li, W.; Risacher, S.L.; et al. Clinical Phenotype of Dementia after Traumatic Brain Injury. J. Neurotrauma 2013, 30, 1117–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. McKee, A.C.; Stein, T.D.; Nowinski, C.J.; Stern, R.A.; Daneshvar, D.H.; Alvarez, V.E.; Lee, H.-S.; Hall, G.; Wojtowicz, S.M.; Baugh, C.M.; et al. The spectrum of disease in chronic traumatic encephalopathy. Brain 2013, 136, 43–64. [Google Scholar] [CrossRef] [PubMed]
  64. de Calignon, A.; Polydoro, M.; Suárez-Calvet, M.; William, C.; Adamowicz, D.H.; Kopeikina, K.J.; Pitstick, R.; Sahara, N.; Ashe, K.H.; Carlson, G.A.; et al. Propagation of Tau Pathology in a Model of Early Alzheimer’s Disease. Neuron 2012, 73, 685–697. [Google Scholar] [CrossRef] [Green Version]
  65. Seeley, W.W.; Crawford, R.K.; Zhou, J.; Miller, B.L.; Greicius, M.D. Neurodegenerative Diseases Target Large-Scale Human Brain Networks. Neuron 2009, 62, 42–52. [Google Scholar] [CrossRef] [Green Version]
  66. Shitaka, Y.; Tran, H.T.; Bennett, R.E.; Sanchez, L.; Levy, M.A.; Dikranian, K.; Brody, D.L. Repetitive Closed-Skull Traumatic Brain Injury in Mice Causes Persistent Multifocal Axonal Injury and Microglial Reactivity. J. Neuropathol. Exp. Neurol. 2011, 70, 551–567. [Google Scholar] [CrossRef]
  67. Johnson, V.E.; Stewart, J.E.; Begbie, F.D.; Trojanowski, J.Q.; Smith, D.H.; Stewart, W. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain 2013, 136, 28–42. [Google Scholar] [CrossRef] [Green Version]
  68. Gentleman, S. Microglia in protein aggregation disorders: Friend or foe? Neuropathol. Appl. Neurobiol. 2013, 39, 45–50. [Google Scholar] [CrossRef] [Green Version]
  69. Holmin, S.; Mathiesen, T. Long-term intracerebral inflammatory response after experimental focal brain injury in rat. Neuroreport 1999, 10, 1889–1891. [Google Scholar] [CrossRef]
  70. Marmarou, A.; Lu, J.; Butcher, I.; McHugh, G.S.; Murray, G.D.; Steyerberg, E.W.; Mushkudiani, N.A.; Choi, S.; Maas, A.I. Prognostic Value of the Glasgow Coma Scale and Pupil Reactivity in Traumatic Brain Injury Assessed Pre-Hospital and on Enrollment: An IMPACT Analysis. J. Neurotrauma 2007, 24, 270–280. [Google Scholar] [CrossRef]
  71. Teasdale, G.; Murray, G.; Parker, L.; Jennett, B. Prognostic Factors in the First Week of Acute Head Injuries. Implication for Treatment. In Proceedings of the 6th European Congress of Neurosurgery, Paris, France, 15–20 July 1979; Springer: Berlin/Heidelberg, Germany, 1979; pp. 13–16. [Google Scholar]
  72. Jennett, B. Defining brain damage after head injury. J. R. Coll. Physicians Lond. 1979, 13, 197. [Google Scholar]
  73. Bhatty, G.B.; Kapoor, N. The Glasgow Coma Scale: A mathematical critique. Acta Neurochir. 1993, 120, 132–135. [Google Scholar] [CrossRef]
  74. Reith, F.C.; Lingsma, H.F.; Gabbe, B.J.; Lecky, F.E.; Roberts, I.; Maas, A.I. Differential effects of the Glasgow Coma Scale Score and its Components: An analysis of 54,069 patients with traumatic brain injury. Injury 2017, 48, 1932–1943. [Google Scholar] [CrossRef]
  75. Teoh, L.S.G.; Gowardman, J.R.; Larsen, P.D.; Green, R.; Galletly, D. Glasgow Coma Scale: Variation in mortality among permutations of specific total scores. Intensiv. Care Med. 2000, 26, 157–161. [Google Scholar] [CrossRef]
  76. McCrea, M.A.; Giacino, J.T.; Barber, J.; Temkin, N.R.; Nelson, L.D.; Levin, H.S.; Dikmen, S.; Stein, M.; Bodien, Y.G.; Boase, K.; et al. Functional Outcomes over the First Year after Moderate to Severe Traumatic Brain Injury in the Prospective, Longitudinal TRACK-TBI Study. JAMA Neurol. 2021, 78, 982–992. [Google Scholar] [CrossRef]
  77. Nelson, L.D.; Temkin, N.R.; Dikmen, S.; Barber, J.; Giacino, J.T.; Yuh, E.; Levin, H.S.; McCrea, M.A.; Stein, M.B.; Mukherjee, P.; et al. Recovery after mild traumatic brain injury in patients presenting to US level I trauma centers: A transforming research and clinical knowledge in traumatic brain injury (TRACK-TBI) study. JAMA Neurol. 2019, 76, 1049–1059. [Google Scholar] [CrossRef]
  78. Nelson, L.D.; Ranson, J.; Ferguson, A.R.; Okonkwo, D.O.; Valadka, A.B.; Manley, G.T.; McCrea, M.A.; The TRACK-TBI Investigators; Riemann, L.; Mikolic, A.; et al. Validating Multi-Dimensional Outcome Assessment Using the Traumatic Brain Injury Common Data Elements: An Analysis of the TRACK-TBI Pilot Study Sample. J. Neurotrauma 2017, 34, 3158–3172. [Google Scholar] [CrossRef]
  79. Kondziella, D.; Friberg, C.K.; Frokjaer, V.G.; Fabricius, M.; Møller, K. Preserved consciousness in vegetative and minimal conscious states: Systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2016, 87, 485–492. [Google Scholar] [CrossRef]
  80. Claassen, J.; Doyle, K.; Matory, A.; Couch, C.; Burger, K.M.; Velazquez, A.; Okonkwo, J.U.; King, J.-R.; Park, S.; Agarwal, S.; et al. Detection of Brain Activation in Unresponsive Patients with Acute Brain Injury. N. Engl. J. Med. 2019, 380, 2497–2505. [Google Scholar] [CrossRef]
  81. Schiff, N.D. Cognitive Motor Dissociation Following Severe Brain Injuries. JAMA Neurol. 2015, 72, 1413–1415. [Google Scholar] [CrossRef]
  82. Seel, R.T.; Sherer, M.; Whyte, J.; Katz, D.I.; Giacino, J.T.; Rosenbaum, A.M.; Hammond, F.M.; Kalmar, K.; Pape, T.L.-B.; Zafonte, R.; et al. Assessment Scales for Disorders of Consciousness: Evidence-Based Recommendations for Clinical Practice and Research. Arch. Phys. Med. Rehabil. 2010, 91, 1795–1813. [Google Scholar] [CrossRef] [PubMed]
  83. Whyte, J.; DiPasquale, M.C.; Vaccaro, M. Assessment of command-following in minimally conscious brain injured patients. Arch. Phys. Med. Rehabil. 1999, 80, 653–660. [Google Scholar] [CrossRef] [PubMed]
  84. Giacino, J.T.; Kalmar, K.; Whyte, J. The JFK Coma Recovery Scale-Revised: Measurement characteristics and diagnostic utility. Arch. Phys. Med. Rehabil. 2004, 85, 2020–2029. [Google Scholar] [CrossRef] [PubMed]
  85. Giacino, J.T.; Fins, J.J.; Laureys, S.; Schiff, N.D. Disorders of consciousness after acquired brain injury: The state of the science. Nat. Rev. Neurol. 2014, 10, 99–114. [Google Scholar] [CrossRef] [PubMed]
  86. Boltzmann, M.; Schmidt, S.B.; Gutenbrunner, C.; Krauss, J.K.; Stangel, M.; Höglinger, G.U.; Wallesch, C.-W.; Rollnik, J.D. The influence of the CRS-R score on functional outcome in patients with severe brain injury receiving early rehabilitation. BMC Neurol. 2021, 21, 44. [Google Scholar] [CrossRef]
  87. Schnakers, C.; Bauer, C.; Formisano, R.; Noé, E.; Llorens, R.; Lejeune, N.; Farisco, M.; Teixeira, L.; Morrissey, A.-M.; De Marco, S.; et al. What names for covert awareness? A systematic review. Front. Hum. Neurosci. 2022, 16, 971315. [Google Scholar] [CrossRef]
  88. Kampfl, A.; Schmutzhard, E.; Franz, G.; Pfausler, B.; Haring, H.-P.; Ulmer, H.; Felber, S.; Golaszewski, S.; Aichner, F. Prediction of recovery from post-traumatic vegetative state with cerebral magnetic-resonance imaging. Lancet 1998, 351, 1763–1767. [Google Scholar] [CrossRef]
  89. Galanaud, D.; Perlbarg, V.; Gupta, R.; Stevens, R.D.; Sanchez, P.; Tollard, E.; De Champfleur, N.M.; Dinkel, J.; Faivre, S.; Stot-Ares, G.; et al. Assessment of White Matter Injury and Outcome in Severe Brain Trauma. A Prospective Multicenter Cohort. Surv. Anesthesiol. 2012, 117, 1300–1310. [Google Scholar] [CrossRef] [Green Version]
  90. Luyt, C.-E.; Galanaud, D.; Perlbarg, V.; Vanhaudenhuyse, A.; Stevens, R.D.; Gupta, R.; Besancenot, H.; Krainik, A.; Audibert, G.; Combes, A.; et al. Diffusion tensor imaging to predict long-term outcome after cardiac arrest: A bicentric pilot study. J. Am. Soc. Anesthesiol. 2012, 117, 1311–1321. [Google Scholar] [CrossRef] [Green Version]
  91. Voss, H.U.; Uluç, A.M.; Dyke, J.P.; Watts, R.; Kobylarz, E.J.; McCandliss, B.D.; Heier, L.A.; Beattie, B.J.; Hamacher, K.A.; Vallabhajosula, S.; et al. Possible axonal regrowth in late recovery from the minimally conscious state. J. Clin. Investig. 2006, 116, 2005–2011. [Google Scholar] [CrossRef] [Green Version]
  92. Bruno, M.-A.; Gosseries, O.; Ledoux, D.; Hustinx, R.; Laureys, S. Assessment of consciousness with electrophysiological and neurological imaging techniques. Curr. Opin. Crit. Care 2011, 17, 146–151. [Google Scholar] [CrossRef]
  93. Sandroni, C.; Cavallaro, F.; Callaway, C.W.; D’arrigo, S.; Sanna, T.; Kuiper, M.A.; Biancone, M.; Della Marca, G.; Farcomeni, A.; Nolan, J.P. Predictors of poor neurological outcome in adult comatose survivors of cardiac arrest: A systematic review and meta-analysis. Part 2: Patients treated with therapeutic hypothermia. Resuscitation 2013, 84, 1324–1338. [Google Scholar] [CrossRef]
  94. Engemann, D.A.; Raimondo, F.; King, J.-R.; Rohaut, B.; Louppe, G.; Faugeras, F.; Annen, J.; Cassol, H.; Gosseries, O.; Fernandez-Slezak, D.; et al. Robust EEG-based cross-site and cross-protocol classification of states of consciousness. Brain 2018, 141, 3179–3192. [Google Scholar] [CrossRef] [Green Version]
  95. Chennu, S.; Annen, J.; Wannez, S.; Thibaut, A.; Chatelle, C.; Cassol, H.; Martens, G.; Schnakers, C.; Gosseries, O.; Menon, D.; et al. Brain networks predict metabolism, diagnosis and prognosis at the bedside in disorders of consciousness. Brain 2017, 140, 2120–2132. [Google Scholar] [CrossRef]
  96. Bagnato, S.; Boccagni, C.; Sant’angelo, A.; Prestandrea, C.; Mazzilli, R.; Galardi, G. EEG predictors of outcome in patients with disorders of consciousness admitted for intensive rehabilitation. Clin. Neurophysiol. 2015, 126, 959–966. [Google Scholar] [CrossRef]
  97. Estraneo, A.; Fiorenza, S.; Magliacano, A.; Formisano, R.; Mattia, D.; Grippo, A.; Romoli, A.M.; Angelakis, E.; Cassol, H.; Thibaut, A.; et al. Multicenter prospective study on predictors of short-term outcome in disorders of consciousness. Neurology 2020, 95, e1488–e1499. [Google Scholar] [CrossRef]
  98. Laureys, S.; Goldman, S.; Phillips, C.; Van Bogaert, P.; Aerts, J.; Luxen, A.; Franck, G.; Maquet, P. Impaired Effective Cortical Connectivity in Vegetative State: Preliminary Investigation Using PET. Neuroimage 1999, 9, 377–382. [Google Scholar] [CrossRef] [Green Version]
  99. Laureys, S.; Lemaire, C.; Maquet, P.; Phillips, C.; Franck, G. Cerebral metabolism during vegetative state and after recovery to consciousness. J. Neurol. Neurosurg. Psychiatry 1999, 67, 121–122. [Google Scholar] [CrossRef] [Green Version]
  100. Phillips, C.L.; Bruno, M.-A.; Maquet, P.; Boly, M.; Noirhomme, Q.; Schnakers, C.; Vanhaudenhuyse, A.; Bonjean, M.; Hustinx, R.; Moonen, G.; et al. “Relevance vector machine” consciousness classifier applied to cerebral metabolism of vegetative and locked-in patients. Neuroimage 2011, 56, 797–808. [Google Scholar] [CrossRef]
  101. Corrigan, J.D.; Smith-Knapp, K.; Granger, C.V. Validity of the functional independence measure for persons with traumatic brain injury. Arch. Phys. Med. Rehabil. 1997, 78, 828–834. [Google Scholar] [CrossRef]
  102. Demertzi, A.; Soddu, A.; Laureys, S. Consciousness supporting networks. Curr. Opin. Neurobiol. 2013, 23, 239–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Boly, M.; Phillips, C.; Balteau, E.; Schnakers, C.; Degueldre, C.; Moonen, G.; Luxen, A.; Peigneux, P.; Faymonville, M.-E.; Maquet, P.; et al. Consciousness and cerebral baseline activity fluctuations. Hum. Brain Mapp. 2008, 29, 868–874. [Google Scholar] [CrossRef] [PubMed]
  104. Demertzi, A.; Vanhaudenhuyse, A.; Brédart, S.; Heine, L.; di Perri, C.; Laureys, S. Looking for the Self in Pathological Unconsciousness. Front. Hum. Neurosci. 2013, 7, 538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Boly, M.; Tshibanda, L.; Vanhaudenhuyse, A.; Noirhomme, Q.; Schnakers, C.; Ledoux, D.; Boveroux, P.; Garweg, C.; Lambermont, B.; Phillips, C.; et al. Functional connectivity in the default network during resting state is preserved in a vegetative but not in a brain dead patient. Hum. Brain Mapp. 2009, 30, 2393–2400. [Google Scholar] [CrossRef] [Green Version]
  106. Vanhaudenhuyse, A.; Noirhomme, Q.; Tshibanda, L.J.-F.; Bruno, M.-A.; Boveroux, P.; Schnakers, C.; Soddu, A.; Perlbarg, V.; Ledoux, D.; Brichant, J.-F.; et al. Default network connectivity reflects the level of consciousness in non-communicative brain-damaged patients. Brain 2010, 133, 161–171. [Google Scholar] [CrossRef] [Green Version]
  107. Cauda, F.; Micon, B.M.; Sacco, K.; Duca, S.; D’Agata, F.; Geminiani, G.; Canavero, S. Disrupted intrinsic functional connectivity in the vegetative state. J. Neurol. Neurosurg. Psychiatry 2009, 80, 429–431. [Google Scholar] [CrossRef] [Green Version]
  108. Di, H.; Boly, M.; Weng, X.; Ledoux, D.; Laureys, S. Neuroimaging activation studies in the vegetative state: Predictors of recovery? Clin. Med. 2008, 8, 502. [Google Scholar] [CrossRef]
  109. Edlow, B.L.; Giacino, J.T.; Wu, O. Functional MRI and outcome in traumatic coma. Curr. Neurol. Neurosci. Rep. 2013, 13, 375. [Google Scholar] [CrossRef] [Green Version]
  110. Graham, N.S.N.; Zimmerman, K.A.; Moro, F.; Heslegrave, A.; Maillard, S.A.; Bernini, A.; Miroz, J.-P.; Donat, C.K.; Lopez, M.Y.; Bourke, N.; et al. Axonal marker neurofilament light predicts long-term outcomes and progressive neurodegeneration after traumatic brain injury. Sci. Transl. Med. 2021, 13, eabg9922. [Google Scholar] [CrossRef]
  111. Czeiter, E.; Amrein, K.; Gravesteijn, B.Y.; Lecky, F.; Menon, D.K.; Mondello, S.; Newcombe, V.F.; Richter, S.; Steyerberg, E.W.; Vyvere, T.V.; et al. Blood biomarkers on admission in acute traumatic brain injury: Relations to severity, CT findings and care path in the CENTER-TBI study. Ebiomedicine 2020, 56, 102785. [Google Scholar] [CrossRef]
  112. Helmrich, I.R.A.R.; Czeiter, E.; Amrein, K.; Büki, A.; Lingsma, H.F.; Menon, D.K.; Mondello, S.; Steyerberg, E.W.; von Steinbüchel, N.; Wang, K.K.W.; et al. Incremental prognostic value of acute serum biomarkers for functional outcome after traumatic brain injury (CENTER-TBI): An observational cohort study. Lancet Neurol. 2022, 21, 792–802. [Google Scholar] [CrossRef]
  113. Okonkwo, D.O.; Puffer, R.C.; Puccio, A.M.; Yuh, E.L.; Yue, J.K.; Diaz-Arrastia, R.; Korley, F.K.; Wang, K.K.W.; Sun, X.; Taylor, S.R.; et al. Point-of-Care Platform Blood Biomarker Testing of Glial Fibrillary Acidic Protein versus S100 Calcium-Binding Protein B for Prediction of Traumatic Brain Injuries: A Transforming Research and Clinical Knowledge in Traumatic Brain Injury Study. J. Neurotrauma 2020, 37, 2460–2467. [Google Scholar] [CrossRef]
  114. Wang, K.K.; Kobeissy, F.H.; Shakkour, Z.; Tyndall, J.A. Thorough overview of ubiquitin C-terminal hydrolase-L1 and glial fibrillary acidic protein as tandem biomarkers recently cleared by US Food and Drug Administration for the evaluation of intracranial injuries among patients with traumatic brain injury. Acute Med. Surg. 2021, 8, e622. [Google Scholar] [CrossRef]
  115. Papa, L.; Silvestri, S.; Brophy, G.M.; Giordano, P.; Falk, J.L.; Braga, C.F.; Tan, C.N.; Ameli, N.J.; Demery, J.A.; Dixit, N.K.; et al. GFAP Out-Performs S100β in Detecting Traumatic Intracranial Lesions on Computed Tomography in Trauma Patients with Mild Traumatic Brain Injury and Those with Extracranial Lesions. J. Neurotrauma 2014, 31, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
  116. Mondello, S.; Jeromin, A.; Buki, A.; Bullock, R.; Czeiter, E.; Kovacs, N.; Barzo, P.; Hayes, R.L.; Wang, K.K.W.; Yang, Z.; et al. Glial Neuronal Ratio: A Novel Index for Differentiating Injury Type in Patients with Severe Traumatic Brain Injury. J. Neurotrauma 2012, 29, 1096–1104. [Google Scholar] [CrossRef] [Green Version]
  117. Mondello, S.; Papa, L.; Buki, A.; Bullock, M.R.; Czeiter, E.; Tortella, F.C.; Wang, K.K.; Hayes, R.L. Neuronal and glial markers are differently associated with computed tomography findings and outcome in patients with severe traumatic brain injury: A case control study. Crit. Care 2011, 15, R156. [Google Scholar] [CrossRef] [Green Version]
  118. Whitehouse, D.P.; Monteiro, M.; Czeiter, E.; Vyvere, T.V.; Valerio, F.; Ye, Z.; Amrein, K.; Kamnitsas, K.; Xu, H.; Yang, Z.; et al. Relationship of admission blood proteomic biomarkers levels to lesion type and lesion burden in traumatic brain injury: A CENTER-TBI study. Ebiomedicine 2022, 75, 103777. [Google Scholar] [CrossRef] [PubMed]
  119. Korley, F.K.; Jain, S.; Sun, X.; Puccio, A.M.; Yue, J.K.; Gardner, R.C.; Wang, K.K.W.; Okonkwo, D.O.; Yuh, E.L.; Mukherjee, P.; et al. Prognostic value of day-of-injury plasma GFAP and UCH-L1 concentrations for predicting functional recovery after traumatic brain injury in patients from the US TRACK-TBI cohort: An observational cohort study. Lancet Neurol. 2022, 21, 803–813. [Google Scholar] [CrossRef]
  120. Seabury, S.A.; Gaudette, É.; Goldman, D.P.; Markowitz, A.J.; Brooks, J.; McCrea, M.A.; Okonkwo, D.O.; Manley, G.T.; The TRACK-TBI Investigators. Assessment of follow-up care after emergency department presentation for mild traumatic brain injury and concussion: Results from the TRACK-TBI study. JAMA Netw. Open 2018, 1, e180210. [Google Scholar] [CrossRef] [PubMed]
  121. Armstrong, M.J.; Giacino, J.T.; Katz, D.I.; Schiff, N.D.; Whyte, J.; Ashman, E.J.; Ashwal, S.; Barbano, R.; Hammond, F.M.; Laureys, S.; et al. Author response: Practice guideline update recommendations summary: Disorders of consciousness: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology; the American Congress of Rehabilitation Medicine; and the National Institute on Disability, Independent Living, and Rehabilitation Research. Neurology 2019, 92, 1699–1709. [Google Scholar] [CrossRef]
  122. Giacino, J.T.; Whyte, J.; Bagiella, E.; Kalmar, K.; Childs, N.; Khademi, A.; Eifert, B.; Long, D.; Katz, D.I.; Cho, S.; et al. Placebo-Controlled Trial of Amantadine for Severe Traumatic Brain Injury. N. Engl. J. Med. 2012, 366, 819–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Eban-Rothschild, A.; Rothschild, G.; Giardino, W.J.; Jones, J.R.; de Lecea, L. VTA dopaminergic neurons regulate ethologically relevant sleep–wake behaviors. Nat. Neurosci. 2016, 19, 1356–1366. [Google Scholar] [CrossRef] [PubMed]
  124. Fridman, E.A.; Beattie, B.J.; Broft, A.; Laureys, S.; Schiff, N.D. Regional cerebral metabolic patterns demonstrate the role of anterior forebrain mesocircuit dysfunction in the severely injured brain. Proc. Natl. Acad. Sci. USA 2014, 111, 6473–6478. [Google Scholar] [CrossRef]
  125. Matsuda, W.; Komatsu, Y.; Yanaka, K.; Matsumura, A. Levodopa treatment for patients in persistent vegetative or minimally conscious states. Neuropsychol. Rehabil. 2005, 15, 414–427. [Google Scholar] [CrossRef]
  126. Ugoya, S.O.; Akinyemi, R.O. The Place of l-Dopa/Carbidopa in Persistent Vegetative State. Clin. Neuropharmacol. 2010, 33, 279–284. [Google Scholar] [CrossRef]
  127. Tucker, C.; Sandhu, K. The Effectiveness of Zolpidem for the Treatment of Disorders of Consciousness. Neurocritical Care 2016, 24, 488–493. [Google Scholar] [CrossRef]
  128. Sutton, J.A.; Clauss, R.P. A review of the evidence of zolpidem efficacy in neurological disability after brain damage due to stroke, trauma and hypoxia: A justification of further clinical trials. Brain Inj. 2017, 31, 1019–1027. [Google Scholar] [CrossRef]
  129. Williams, S.T.; Conte, M.M.; Goldfine, A.M.; Noirhomme, Q.; Gosseries, O.; Thonnard, M.; Beattie, B.; Hersh, J.; Katz, D.I.; Victor, J.D.; et al. Common resting brain dynamics indicate a possible mechanism underlying zolpidem response in severe brain injury. eLife 2013, 2, e01157. [Google Scholar] [CrossRef]
  130. Whyte, J.; Rajan, R.; Rosenbaum, A.; Katz, D.; Kalmar, K.; Seel, R.; Greenwald, B.; Zafonte, R.; Demarest, D.; Brunner, R.; et al. Zolpidem and restoration of consciousness. Am. J. Phys. Med. Rehabil. 2014, 93, 101–113. [Google Scholar] [CrossRef]
  131. Thonnard, M.; Gosseries, O.; Demertzi, A.; Lugo, Z.; Vanhaudenhuyse, A.; Bruno, M.-A.; Chatelle, C.; Thibaut, A.; Charland-Verville, V.; Habbal, D.; et al. Effect of zolpidem in chronic disorders of consciousness: A prospective open-label study. Funct. Neurol. 2013, 28, 259. [Google Scholar]
  132. Lin, P.-H.; Kuo, L.-T.; Luh, H.-T. The Roles of Neurotrophins in Traumatic Brain Injury. Life 2021, 12, 26. [Google Scholar] [CrossRef] [PubMed]
  133. Schoch, K.M.; Madathil, S.K.; Saatman, K.E. Genetic Manipulation of Cell Death and Neuroplasticity Pathways in Traumatic Brain Injury. Neurotherapeutics 2012, 9, 323–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Stein, D.G.; Hoffman, S.W. Concepts of CNS Plasticity in the Context of Brain Damage and Repair. J. Head Trauma Rehabil. 2003, 18, 317–341. [Google Scholar] [CrossRef] [PubMed]
  135. Yu, T.-S.; Zhang, G.; Liebl, D.J.; Kernie, S.G. Traumatic Brain Injury-Induced Hippocampal Neurogenesis Requires Activation of Early Nestin-Expressing Progenitors. J. Neurosci. 2008, 28, 12901–12912. [Google Scholar] [CrossRef] [Green Version]
  136. Madathil, S.K.; Saatman, K.E. IGF-1/IGF-R Signaling in Traumatic Brain Injury: Impact on Cell Survival, Neurogenesis, and Behavioral Outcome; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2015. [Google Scholar]
  137. Skinner, S.J.; Geaney, M.S.; Rush, R.; Rogers, M.-L.; Emerich, D.F.; Thanos, C.G.; Vasconcellos, A.V.; Tan, P.L.; Elliott, R.B. Choroid plexus transplants in the treatment of brain diseases. Xenotransplantation 2006, 13, 284–288. [Google Scholar] [CrossRef]
  138. Johanson, C.; Stopa, E.; Baird, A.; Sharma, H. Traumatic brain injury and recovery mechanisms: Peptide modulation of periventricular neurogenic regions by the choroid plexus–CSF nexus. J. Neural Transm. 2011, 118, 115–133. [Google Scholar] [CrossRef] [Green Version]
  139. Skinner, S.J.M.; Geaney, M.S.; Lin, H.; Muzina, M.; Anal, A.K.; Elliott, R.B.; Tan, P.L.J. Encapsulated living choroid plexus cells: Potential long-term treatments for central nervous system disease and trauma. J. Neural Eng. 2009, 6, 065001. [Google Scholar] [CrossRef]
  140. Pedraza, C.E.; Podlesniy, P.; Vidal, N.; Arévalo, J.C.; Lee, R.; Hempstead, B.; Ferrer, I.; Iglesias, M.; Espinet, C. Pro-NGF isolated from the human brain affected by Alzheimer’s disease induces neuronal apoptosis mediated by p75NTR. Am. J. Pathol. 2005, 166, 533–543. [Google Scholar] [CrossRef]
  141. Harrington, A.W.; Leiner, B.; Blechschmitt, C.; Arevalo, J.C.; Lee, R.; Mörl, K.; Meyer, M.; Hempstead, B.L.; Yoon, S.O.; Giehl, K.M. Secreted proNGF is a pathophysiological death-inducing ligand after adult CNS injury. Proc. Natl. Acad. Sci. USA 2004, 101, 6226–6230. [Google Scholar] [CrossRef]
  142. Chiaretti, A.; Antonelli, A.; Riccardi, R.; Genovese, O.; Pezzotti, P.; Di Rocco, C.; Tortorolo, L.; Piedimonte, G. Nerve growth factor expression correlates with severity and outcome of traumatic brain injury in children. Eur. J. Paediatr. Neurol. 2008, 12, 195–204. [Google Scholar] [CrossRef] [Green Version]
  143. Munoz, M.J.; Kumar, R.G.; Oh, B.-M.; Conley, Y.P.; Wang, Z.; Failla, M.D.; Wagner, A.K. Cerebrospinal Fluid Cortisol Mediates Brain-Derived Neurotrophic Factor Relationships to Mortality after Severe TBI: A Prospective Cohort Study. Front. Mol. Neurosci. 2017, 10, 44. [Google Scholar] [CrossRef] [Green Version]
  144. Bliźniewska-Kowalska, K.; Łukasik, M.; Gałecki, P. Cerebrolysin—Mechanism of action and application in psychiatry and neurology. Pharmacother. Psychiatry Neurol. 2019, 35, 9–23. [Google Scholar] [CrossRef]
  145. König, P.; Waanders, R.; Witzmann, A.; Lanner, G.; Haffner, Z.; Haninec, P.; Gmeinbauer, R.; Zimmermann-Meinzingen, S. Cerebrolysin in traumatic brain injury—A pilot study of a neurotrophic and neurogenic agent in the treatment of acute traumatic brain injury. J. Neurol. Neurochir. Psychiatr. 2006, 7, 12–20. [Google Scholar]
  146. Chen, C.-C.; Wei, S.-T.; Tsaia, S.-C.; Chen, X.-X.; Cho, D.-Y. Cerebrolysin enhances cognitive recovery of mild traumatic brain injury patients: Double-blind, placebo-controlled, randomized study. Br. J. Neurosurg. 2013, 27, 803–807. [Google Scholar] [CrossRef]
  147. Muresanu, D.F.; Ciurea, A.V.; Gorgan, R.M.; Gheorghita, E.; Florian, S.I.; Stan, H.; Blaga, A.; Ianovici, N.; Iencean, S.M.; Turliuc, D.; et al. A retrospective, multi-center cohort study evaluating the severity-related effects of cerebrolysin treatment on clinical outcomes in traumatic brain injury. CNS Neurol. Disord. Drug Targets 2015, 14, 587–599. [Google Scholar] [CrossRef] [Green Version]
  148. Lefaucheur, J.-P.; André-Obadia, N.; Antal, A.; Ayache, S.S.; Baeken, C.; Benninger, D.H.; Cantello, R.M.; Cincotta, M.; de Carvalho, M.; De Ridder, D.; et al. Evidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS). Clin. Neurophysiol. 2014, 125, 2150–2206. [Google Scholar] [CrossRef]
  149. Cavinato, M.; Genna, C.; Formaggio, E.; Gregorio, C.; Storti, S.F.; Manganotti, P.; Casanova, E.; Piperno, R.; Piccione, F. Behavioural and electrophysiological effects of tDCS to prefrontal cortex in patients with disorders of consciousness. Clin. Neurophysiol. 2019, 130, 231–238. [Google Scholar] [CrossRef]
  150. Ali, M.M.; Sellers, K.K.; Fröhlich, F. Transcranial Alternating Current Stimulation Modulates Large-Scale Cortical Network Activity by Network Resonance. J. Neurosci. 2013, 33, 11262–11275. [Google Scholar] [CrossRef]
  151. Helfrich, R.F.; Schneider, T.R.; Rach, S.; Trautmann-Lengsfeld, S.A.; Engel, A.K.; Herrmann, C.S. Entrainment of Brain Oscillations by Transcranial Alternating Current Stimulation. Curr. Biol. 2014, 24, 333–339. [Google Scholar] [CrossRef] [Green Version]
  152. Thibaut, A.; Bruno, M.-A.; LeDoux, D.; Demertzi, A.; Laureys, S. tDCS in patients with disorders of consciousness: Sham-controlled randomized double-blind study. Neurology 2014, 82, 1112–1118. [Google Scholar] [CrossRef]
  153. Martens, G.; Lejeune, N.; O’Brien, A.T.; Fregni, F.; Martial, C.; Wannez, S.; Laureys, S.; Thibaut, A. Randomized controlled trial of home-based 4-week tDCS in chronic minimally conscious state. Brain Stimul. 2018, 11, 982–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Müri, R.M.; Cazzoli, D.; Nef, T.; Mosimann, U.P.; Hopfner, S.; Nyffeler, T. Non-Invasive Brain Stimulation in Neglect Rehabilitation: An Update. Front. Hum. Neurosci. 2013, 7, 248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Cincotta, M.; Giovannelli, F.; Chiaramonti, R.; Bianco, G.; Godone, M.; Battista, D.; Cardinali, C.; Borgheresi, A.; Sighinolfi, A.; D’Avanzo, A.M.; et al. No effects of 20 Hz-rTMS of the primary motor cortex in vegetative state: A randomised, sham-controlled study. Cortex 2015, 71, 368–376. [Google Scholar] [CrossRef] [PubMed]
  156. Liu, P.; Gao, J.; Pan, S.; Meng, F.; Pan, G.; Li, J.; Luo, B. Effects of High-Frequency Repetitive Transcranial Magnetic Stimulation on Cerebral Hemodynamics in Patients with Disorders of Consciousness: A Sham-Controlled Study. Eur. Neurol. 2016, 76, 1–7. [Google Scholar] [CrossRef] [PubMed]
  157. Naro, A.; Russo, M.; Leo, A.; Bramanti, P.; Quartarone, A.; Calabrò, R.S. A single session of repetitive transcranial magnetic stimulation over the dorsolateral prefrontal cortex in patients with unresponsive wakefulness syndrome: Preliminary results. Neurorehabilit. Neural Repair 2015, 29, 603–613. [Google Scholar] [CrossRef]
  158. Casali, A.G.; Gosseries, O.; Rosanova, M.; Boly, M.; Sarasso, S.; Casali, K.R.; Casarotto, S.; Bruno, M.-A.; Laureys, S.; Tononi, G.; et al. A theoretically based index of consciousness independent of sensory processing and behavior. Sci. Transl. Med. 2013, 5, 198ra105. [Google Scholar] [CrossRef]
  159. Xia, X.; Wang, Y.; Li, C.; Li, X.; He, J.; Bai, Y. Transcranial magnetic stimulation-evoked connectivity reveals modulation effects of repetitive transcranial magnetic stimulation on patients with disorders of consciousness. Neuroreport 2019, 30, 1307–1315. [Google Scholar] [CrossRef]
  160. Mensen, A.; Bodart, O.; Thibaut, A.; Wannez, S.; Annen, J.; Laureys, S.; Gosseries, O. Decreased Evoked Slow-Activity after tDCS in Disorders of Consciousness. Front. Syst. Neurosci. 2020, 14, 62. [Google Scholar] [CrossRef]
  161. Blethyn, K.L.; Hughes, S.W.; Crunelli, V. Evidence for electrical synapses between neurons of the nucleus reticularis thalami in the adult brain in vitro. Thalamus Relat. Syst. 2008, 4, 13–20. [Google Scholar] [CrossRef] [Green Version]
  162. Hindriks, R.; van Putten, M.J. Thalamo-cortical mechanisms underlying changes in amplitude and frequency of human alpha oscillations. Neuroimage 2013, 70, 150–163. [Google Scholar] [CrossRef]
  163. Cooper, J.B.; Jane, J.A.; Alves, W.M.; Cooper, E.B. Right median nerve electrical stimulation to hasten awakening from coma. Brain Inj. 1999, 13, 261–267. [Google Scholar] [CrossRef]
  164. Peri, C.V.; Shaffrey, M.E.; Farace, E.; Cooper, E.; Alves, W.M.; Cooper, J.B.; Young, J.S.; Jane, J.A. Pilot study of electrical stimulation on median nerve in comatose severe brain injured patients: 3-month outcome. Brain Inj. 2001, 15, 903–910. [Google Scholar] [CrossRef]
  165. Lei, J.; Wang, L.; Gao, G.; Cooper, E.; Jiang, J.; Hånell, A.; Greer, J.E.; Jacobs, K.M.; Guilfoyle, M.R.; Carpenter, K.L.; et al. Right Median Nerve Electrical Stimulation for Acute Traumatic Coma Patients. J. Neurotrauma 2015, 32, 1584–1589. [Google Scholar] [CrossRef]
  166. Edlow, B.L.; Sanz, L.R.D.; Polizzotto, L.; Pouratian, N.; Rolston, J.D.; Snider, S.B.; Thibaut, A.; Stevens, R.D.; Gosseries, O.; Akbari, Y.; et al. Therapies to Restore Consciousness in Patients with Severe Brain Injuries: A Gap Analysis and Future Directions. Neurocritical Care 2021, 35, 68–85. [Google Scholar] [CrossRef]
  167. Iriki, A.; Corazzol, M.; Lio, G.; Lefevre, A.; Deiana, G.; Tell, L.; André-Obadia, N.; Bourdillon, P.; Guenot, M.; Desmurget, M.; et al. Faculty Opinions recommendation of Restoring consciousness with vagus nerve stimulation. Curr. Biol. 2017, 27, R994–R996. [Google Scholar] [CrossRef]
  168. Hakon, J.; Moghiseh, M.; Poulsen, I.; Msc, C.M.; Hansen, C.P.; Sabers, A. Transcutaneous Vagus Nerve Stimulation in Patients with Severe Traumatic Brain Injury: A Feasibility Trial. Neuromodul. Technol. Neural Interface 2020, 23, 859–864. [Google Scholar] [CrossRef]
  169. Noé, E.; Ferri, J.; Colomer, C.; Moliner, B.; O’Valle, M.; Ugart, P.; Rodriguez, C.; Llorens, R. Feasibility, safety and efficacy of transauricular vagus nerve stimulation in a cohort of patients with disorders of consciousness. Brain Stimul. 2020, 13, 427–429. [Google Scholar] [CrossRef] [Green Version]
  170. Giacino, J.T. Sensory stimulation: Theoretical perspectives and the evidence for effectiveness. NeuroRehabilitation 1996, 6, 69–78. [Google Scholar] [CrossRef]
  171. O’kelly, J.; James, L.; Palaniappan, R.; Taborin, J.; Fachner, J.; Magee, W.L. Neurophysiological and Behavioral Responses to Music Therapy in Vegetative and Minimally Conscious States. Front. Hum. Neurosci. 2013, 7, 884. [Google Scholar] [CrossRef] [Green Version]
  172. Perrin, F.; Castro, M.; Tillmann, B.; Luauté, J. Promoting the use of personally relevant stimuli for investigating patients with disorders of consciousness. Front. Psychol. 2015, 6, 1102. [Google Scholar] [CrossRef] [Green Version]
  173. Boltzmann, M.; Schmidt, S.B.; Gutenbrunner, C.; Krauss, J.K.; Stangel, M.; Höglinger, G.U.; Wallesch, C.-W.; Münte, T.F.; Rollnik, J.D. Auditory Stimulation Modulates Resting-State Functional Connectivity in Unresponsive Wakefulness Syndrome Patients. Front. Neurosci. 2021, 15, 554194. [Google Scholar] [CrossRef] [PubMed]
  174. Li, X.; Li, C.; Hu, N.; Wang, T. Music Interventions for Disorders of Consciousness: A Systematic Review and Meta-analysis. J. Neurosci. Nurs. 2020, 52, 146–151. [Google Scholar] [CrossRef] [PubMed]
  175. Frazzitta, G.; Zivi, I.; Valsecchi, R.; Bonini, S.; Maffia, S.; Molatore, K.; Sebastianelli, L.; Zarucchi, A.; Matteri, D.; Ercoli, G.; et al. Effectiveness of a very early stepping verticalization protocol in severe acquired brain injured patients: A randomized pilot study in ICU. PLoS ONE 2016, 11, e0158030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Cantor, J.B.; Ashman, T.; Bushnik, T.; Cai, X.; Farrell-Carnahan, L.; Gumber, S.; Hart, T.; Rosenthal, J.; Dijkers, M. Systematic review of interventions for fatigue after traumatic brain injury: A NIDRR traumatic brain injury model systems study. J. Head Trauma Rehabil. 2014, 29, 490–497. [Google Scholar] [CrossRef]
  177. Winward, C.; Sackley, C.; Metha, Z.; Rothwell, P.M. A Population-Based Study of the Prevalence of Fatigue after Transient Ischemic Attack and Minor Stroke. Stroke 2009, 40, 757–761. [Google Scholar] [CrossRef] [Green Version]
  178. Ali, A.M.; Morfin, J.B.; Mills, J.A.; Pasipanodya, E.C.; Maas, Y.J.M.; Huang, E.; Dirlikov, B.M.; Englander, J.; Zedlitz, A. Fatigue after Traumatic Brain Injury: A Systematic Review. J. Head Trauma Rehabil. 2022, 37, E249–E257. [Google Scholar] [CrossRef]
  179. Johansson, B.; Bjuhr, H.; Rönnbäck, L. Mindfulness-based stress reduction (MBSR) improves long-term mental fatigue after stroke or traumatic brain injury. Brain Inj. 2012, 26, 1621–1628. [Google Scholar] [CrossRef]
  180. Edlow, B.L.; Claassen, J.; Schiff, N.D.; Greer, D.M. Recovery from disorders of consciousness: Mechanisms, prognosis and emerging therapies. Nat. Rev. Neurol. 2021, 17, 135–156. [Google Scholar] [CrossRef]
  181. Schnakers, C.; Monti, M.M. Disorders of consciousness after severe brain injury: Therapeutic options. Curr. Opin. Neurol. 2017, 30, 573–579. [Google Scholar] [CrossRef]
  182. Thibaut, A.; Schiff, N.; Giacino, J.; Laureys, S.; Gosseries, O. Therapeutic interventions in patients with prolonged disorders of consciousness. Lancet Neurol. 2019, 18, 600–614. [Google Scholar] [CrossRef]
  183. Andreas Bender, S.B.; Bodechtel, U.; Eifert, B.; Elsner, B.; Feddersen, B.; Freivogel, S.; Hoffmann, B.; Hucke, B.; Huge, V.; HundGeorgiadis, M.; et al. S3-LL Neurologische Rehabilitation bei Koma und schwerer Bewusstseinsstörung im Erwachsenenalter. In Leitlinien für die Neurorehabilitation, 1st ed.; Deutsche Gesellschaft für Neurorehabilitation e.v. (Dgnr) (Hrsgb.): Rheinbach, Germany, 2022. [Google Scholar]
  184. Kondziella, D.; Bender, A.; Diserens, K.; van Erp, W.; Estraneo, A.; Formisano, R.; Laureys, S.; Naccache, L.; Ozturk, S.; Rohaut, B.; et al. European Academy of Neurology guideline on the diagnosis of coma and other disorders of consciousness. Eur. J. Neurol. 2020, 27, 741–756. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The CRS-R improves assessment of minimal cognitive competencies of patients with severe DoC. The scale uses modality-specific items (auditory, visual, motor, oromotor, and communicative responses) and items related to the patient’s activation level. Due to the good item operationalization, the high reliability, validity, and sensitivity compared to the Glasgow Coma Scale (GCS), the CRS-R scale is widely used and internationally recognized.
Figure 1. The CRS-R improves assessment of minimal cognitive competencies of patients with severe DoC. The scale uses modality-specific items (auditory, visual, motor, oromotor, and communicative responses) and items related to the patient’s activation level. Due to the good item operationalization, the high reliability, validity, and sensitivity compared to the Glasgow Coma Scale (GCS), the CRS-R scale is widely used and internationally recognized.
Ctn 07 00021 g001
Figure 2. Scheme of the mesocircuit model adapted by Giacino, 2014. The model represents the key anatomical structures for stimulating therapies in severe DoC. The mesocircuit model integrates neural components that maintain consciousness. The thalamus plays a central role and is activated via a cortico-thalamic loop, in particular via frontal cortex areas, and is inhibited and modulated by inhibitory signals e.g., from the globus pallidus internus. The thalamus activates other cortex areas via feedback loops. In structural brain damage, particularly when neurons in the central thalamus are damaged and afferent input to striatal neurons is lost, inhibitory signals from the striatum to the globus pallidus are inhibited, resulting in an overstimulated pallido-thalamic inhibition which decreases thalamic activity. GABAA α-1 subunit is typically abundantly expressed within the globus pallidus internus. The administration of zolpidem may inhibit the latter, thereby replacing its inhibitory role originating from the striatum. Consequently, this process could lead to an increase in thalamic excitatory modulation on prefrontal cortices.
Figure 2. Scheme of the mesocircuit model adapted by Giacino, 2014. The model represents the key anatomical structures for stimulating therapies in severe DoC. The mesocircuit model integrates neural components that maintain consciousness. The thalamus plays a central role and is activated via a cortico-thalamic loop, in particular via frontal cortex areas, and is inhibited and modulated by inhibitory signals e.g., from the globus pallidus internus. The thalamus activates other cortex areas via feedback loops. In structural brain damage, particularly when neurons in the central thalamus are damaged and afferent input to striatal neurons is lost, inhibitory signals from the striatum to the globus pallidus are inhibited, resulting in an overstimulated pallido-thalamic inhibition which decreases thalamic activity. GABAA α-1 subunit is typically abundantly expressed within the globus pallidus internus. The administration of zolpidem may inhibit the latter, thereby replacing its inhibitory role originating from the striatum. Consequently, this process could lead to an increase in thalamic excitatory modulation on prefrontal cortices.
Ctn 07 00021 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lippert, J.; Guggisberg, A.G. Diagnostic and Therapeutic Approaches in Neurorehabilitation after Traumatic Brain Injury and Disorders of Consciousness. Clin. Transl. Neurosci. 2023, 7, 21. https://doi.org/10.3390/ctn7030021

AMA Style

Lippert J, Guggisberg AG. Diagnostic and Therapeutic Approaches in Neurorehabilitation after Traumatic Brain Injury and Disorders of Consciousness. Clinical and Translational Neuroscience. 2023; 7(3):21. https://doi.org/10.3390/ctn7030021

Chicago/Turabian Style

Lippert, Julian, and Adrian G. Guggisberg. 2023. "Diagnostic and Therapeutic Approaches in Neurorehabilitation after Traumatic Brain Injury and Disorders of Consciousness" Clinical and Translational Neuroscience 7, no. 3: 21. https://doi.org/10.3390/ctn7030021

Article Metrics

Back to TopTop