IMR Press / JIN / Volume 22 / Issue 6 / DOI: 10.31083/j.jin2206142
Open Access Review
Updates on Improving Imaging Modalities for Traumatic Brain Injury
Show Less
1 Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
*Correspondence: Brandon.Lucke-Wold@neurosurgery.ufl.edu (Brandon Lucke-Wold)
J. Integr. Neurosci. 2023, 22(6), 142; https://doi.org/10.31083/j.jin2206142
Submitted: 27 April 2023 | Revised: 13 June 2023 | Accepted: 25 June 2023 | Published: 23 October 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The Center for Disease Control and Prevention reports that traumatic brain injury (TBI) was related to over 64,000 deaths in the United States in 2020, equating to more than 611 TBI-related hospitalizations and 176 TBI-related deaths per day. There are both long- and short-term sequelae involved with the pathophysiology of TBI that can range from mild to severe. Recently, more effort has been devoted to understanding the long-term consequences of TBI and how early detection of these injuries can prevent late clinical manifestations. Obtaining proper, detailed imaging is key to guiding the direction of intervention, but there is a gap in the understanding of how TBI imaging can be used to predict and prevent the long-term morbidities seen with even mild forms of TBI. There have been significant strides in the advancement of TBI imaging that allows for quicker, more affordable, and more effective imaging of intracranial bleeds, axonal injury, tissue damage, and more. Despite this, there is still room for improved standardization and more data supporting the justification of using certain imaging modalities. This review aims to outline recent advancements in TBI imaging and areas that require further investigation to improve patient outcomes and minimize the acute and chronic comorbidities associated with TBI.

Keywords
multimodality imaging
diffused neuronal injury
ultra-high field magnetic resonance imaging susceptibility-weighted imaging
diffusion tensor imaging
traumatic microbleeds
convolutional neural networks
perfusion computed tomography
functional magnetic resonance imaging
near-infrared spectroscopy
transcranial doppler
1. Introduction

To prevent the high rates of mortality and long term morbidities associated with traumatic brain injury (TBI), when a patient is admitted to the neuro-critical care unit for TBI there are recommendations for which imaging modalities to proceed with, relevant thresholds, and ranges based on clinical evidence, facilitated primarily by the Brain Trauma Foundation (BTF) [1, 2] and the Seattle International Severe Traumatic Brain Injury Consensus Conferences (SIBICC) [3]. Despite this, the heterogeneity of TBI and associated gaps in the understanding of its pathophysiology hinder reaching a consensus on standardized imaging protocols and classification. The standardization of TBI management protocols has been shown to be an important factor in contributing to lower risk-adjusted in-hospital mortality and further improving clinical outcomes of patients admitted to neuro-critical care units [4]. However, there are practices common in neuro-critical care units that are commonplace, but are not currently supported by the BTF and SIBICC due to a lack of evidence regarding efficacy, and inadequate understanding of how certain parameters affect patient outcomes and how multiple imaging modalities can be used in combination for optimized multimodal monitoring [5].

Multimodality imaging is critical to understanding the numerous, simultaneous physiochemical properties associated with TBI, and can be used to detect pathological changes more effectively and better deliver targeted therapies to specific neural regions. Currently, multimodality monitoring occurs primarily in neuro-critical care units and considers several metrics using both invasive and non-invasive imaging techniques. There are centers that have combined intracranial pressure (ICP), cerebral microdialysis (CMD), and tissue oxygenation for years, but the level of evidence that directs the accompanying thresholds and evidence of the prognostic capabilities of combining these modalities is lacking [6]. The Brain Trauma Foundation provides official guidelines for some of these metrics and thresholds as well, but there is still a lack of evidence to support the parameters associated with several of the techniques used in multimodality imaging [7].

Immediately upon injury, TBI is associated with prolonged inflammatory cytokine upregulation, decreased oligodendrocyte numbers, increased nitric oxide, and reduced cerebral blood flow. These mechanisms, while protective, exacerbate tissue damage by mobilizing immune and glial cells that cause edema, inflammation, and further diffuse damage [8]. The Glasgow Coma Scale (GCS) is the traditional way to diagnose and classify TBI based on neurological responsiveness and has a strong correlation with patient morbidity and mortality [9]. The GCS is based on three major scores that are categorically divided into eye-opening, verbal, and motor capabilities, scaled from 1–6 for a total score from 3–15. The score can then be related to the level of injury, with 3–8 corresponding with a severe injury, 9–12 with a moderate injury, and 13–15 with a mild injury. A multi-center investigation called the Transforming Research and Clinical Knowledge in TBI (TRACK-TBI) assessed the usefulness in using the GCS to inform clinical decision making and found that considering the three categorical scores individually could improve the assessment and treatment of patients with TBI [10]. Furthermore, while the GCS has proven useful in triaging patients and directing treatment, it is unable to exclude long-term consequences, such as post-concussive syndrome, functional outcomes, and overall morbidity [11, 12].

A major goal in the advancement of TBI imaging is to develop techniques that can prognosticate potentially debilitating, chronic consequences of even mild TBI so that early treatment can be implemented to prevent this. The standard initial imaging protocol for TBI involves computed tomography (CT) followed by 1.5 T and 3.0 T magnetic resonance imaging (MRI), often paired with CT angiography to detect any possibility of cerebrovascular injury. CT are relatively quick and affordable and thus are beneficial for triage, time-sensitive decision making, follow-up imaging, and detecting fractures associated with epidural hematomas, vascular injuries, and cerebrospinal fluid leaks [12]. MRI are more expensive and require specialized equipment that may not be accessible in every clinical environment, but are more specific for detecting axonal injuries and pathological blood byproducts following the initial injury. In addition to accessibility and affordability, MRI speed and sensitivity to disruptive motion are also limitations associated with its use [13].

2. Detecting Microbleeds and Diffused Axonal Injury

Minutes to days following TBI, secondary injury can result from excitatory neurotransmitter release, leading to elevated intracellular calcium which activates caspases and free radicals that contribute to tissue degradation and cellular apoptosis [14]. Ultra-high field MRI susceptibility-weighted imaging (SWI) is being investigated as a superior method compared with the traditional evaluation using 1.5 T and 3.0 T MRI, and has been shown to better detect diffuse axonal injury (DAI), which can allow for better prognostication and treatment. SWI MRI combines filtered-phase data and magnitude data gathered from three-dimensional (3D) gradient-echo sequence evaluations to compare the magnetic susceptibility of adjacent tissues and detect early microhemorrhages that may be associated with DAI [5]. Axonal degeneration can be found in as many as 72% of patients with moderate or severe TBI and is related to both the acute clinical manifestation of TBI and progressive, chronic neurodegenerative issues following the initial injury [15, 16, 17]. Traumatic microbleeds result from damage to cerebral vessels and can be used as an indirect marker of DAI [18]. Some studies suggest that this relationship between microbleeds and DAI is questionable and should be further investigated to reliably detect DAI following TBI [19, 20, 21, 22]. Nonetheless, one study shows that the number of microbleeds has a substantial association with the acute clinical state of patients and chronic neurobehavioral parameters following head injury; therefore, obtaining imaging that can reliably detect microbleeds is an important consideration for both the short and long-term benefit of patients [23, 24].

Diffusion tensor imaging (DTI) is another modality that may help advance the classification system for TBI through heightened sensitivity for detecting axonal injuries with more accuracy. DTI uses the spatial diffusion weight of water to determine multiple different TBI parameters, including fractional anisotropy and mean diffusivity [25]. There is a lack of conclusive data on the usefulness of DTI due to the variable pathophysiology and severity of TBI [26]. In general, increases in mean diffusivity and decreases in fractional anisotropy are associated with the decreased structural integrity of neural white matter. These changes can also be seen with other comorbidities and vary between demographics, so useful quantitative assessment via DTI requires a comparison with control readings [27]. In mild TBI, DTI has been shown to detect increases in fractional anisotropy and decreases in diffusivity, which may be associated with acute cytotoxic edema [28]. Some studies suggest that mild TBI patients with severely reduced fractional anisotropy are associated with worse outcomes, as measured by the Glasgow Outcome Scale [29]. Conversely, other studies show that patients with severe TBI and high fractional anisotropy are associated with more favorable outcomes, possibly due to late axonal regrowth [30]. For moderate to severe TBI, fractional anisotropy and diffusivity, as measured by DTI, has been shown to change up to 18 months after the initial injury and may be correlated with long-term functional outcomes [31]. Further work is required to understand the normal range of DTI metrics within different demographics and how these changes correspond with the severity of injury and symptom resolution, and to further standardize DTI interpretation [5, 32]. To better understand the baseline range of normal DTI measurements, acquisition of pre-injury data needs to be improved. This is difficult to do based on the unpredictability of sustaining a TBI; however, one study that was able to image a TBI patient 12 and 23 months prior to the injury and then 2 weeks and 8 months post-injury found that using 7 Tesla MRI was beneficial in collecting this longitudinal data [33].

3. Detecting Tissue Perfusion and Brain Activity

When a mechanical force causes a head injury that results in brain swelling, the increased intracranial mass results in a decrease in intracranial cerebrospinal fluid and blood flow to compensate for the increase in pressure, as described in the Monro-Kellie hypothesis [34]. Initially upon injury, the expansion of the brain is offset by the elastic nature of the brain tissue, but as intracranial pressure increases the compliance of the tissue decreases, creating a pressure gradient that affects cerebral perfusion. Based on these physiological principles, the Lund concept was developed as the first holistic guideline for treating TBI based on brain volume maintenance and optimizing brain perfusion. The Lund concept was followed by alternative guidelines suggested by the BTF based on meta-analyses and systematic reviews. When comparing the Lund concept and BTF guidelines, they differ substantially [35]. The Lund concept suggests a range of 50–70 mmHg while the 2017 BTF and SIBICC guidelines recommend 60–70 mmHg [28, 35]. The lack of consensus between standard practices can lead to differing health outcomes and it is necessary to stay up to date with the evolution of TBI imaging care.

Historically, global cerebral hypoperfusion is associated with worse outcomes and cannot be detected by traditional non-contrast head CT [36]. Tissue perfusion is not only a major concern during the acute phase of TBI, but is linked to chronic clinical deterioration and worsened outcomes following treatment of the primary injury [37]. Tissue perfusion is commonly evaluated with perfusion computed tomography (CTP), dynamic susceptibility-weighted contrast-enhanced perfusion magnetic resonance imaging (DSC-MRI), and MRI arterial spin labeling (ASL) [5, 38]. CTP has been shown to detect cerebral contusions 7 days earlier than non-contrast CT and predict 6-month outcomes through the evaluation of frontal lobe perfusion [39, 40]. CTP has also been shown to be useful in tracking changes in cerebral perfusion pressure and targeting cerebral loci at risk of hypoxia [41]. Despite this, clinical trials that clearly suggest CTP should be part of the standard of care for TBI imaging are lacking [5].

TBI imaging modalities can be categorized as invasive or non-invasive. Non-invasive monitoring modalities are gaining traction in the detection and treatment of TBI-related injuries, but there is still work to be done regarding the standardization and understanding of their benefits, as much of the research originates from single-center retrospective reviews [7]. Invasive approaches are used in iatrogenic hemorrhages in around 10% of cases and are not typically associated with severe, long-term negative effects [42]. Intracranial pressure (ICP) monitoring and cerebral microdialysis (CMD) are two of the primary invasive methods of brain monitoring used following TBI.

ICP monitoring can be used to derive cerebral perfusion pressure and pressure reactivity indices that can provide information about brain tissue oxygenation. The standard practice of ICP involves implementing a closed external ventricular drain, which is affordable and can simultaneously alleviate cerebrospinal fluid (CSF) build-up contributing to elevated ICP, although this carries a higher procedural risk [43]. Due to the risk of inserting closed external ventricular drains, intraparenchymal monitoring devices are becoming more commonplace because they are easier to insert and can be delivered at the patient’s bedside, although they are unable to provide the same inherent CSF drainage utility [42]. A recent advancement in the utilization of intraparenchymal monitoring devices involves their combination with CSF pumps to match the benefit of closed external ventricular drains [44].

CMD can directly measure biomarkers in cerebral extracellular fluid, such as glucose, lactate, and pyruvate, by inserting a catheter into the brain parenchyma [44, 45]. Some studies show that high lactate to pyruvate ratios, low extracellular glucose, low tissue oxygenation, and impaired pressure reactivity indices measured by CMD are associated with worse outcomes [46, 47]. While there is a consensus of appropriate thresholds and management strategies for these modalities based on international consortiums, high-level, evidence-based studies to support these recommendations are lacking [7].

Functional magnetic resonance imaging (fMRI) is useful for measuring the activity of specific areas of neural tissue based on oxygenation [48, 49]. Severe TBI can be associated with less brain tissue functionality which can lead to comatose states and disorders of consciousness [50]. There is a lack of imaging modalities that can directly track and predict the return of consciousness, though fMRI has shown changes in cortical function in unresponsive patients and may represent a promising tool for predicting the prognosis of TBI-associated loss of consciousness and coma [51]. Proton magnetic resonance spectroscopy (1H-MRS) is another imaging modality that can be useful in prognosticating the functional outcomes of TBI patients. 1H-MRS detects the interaction of protons to quantify the cellular changes in neural tissue based on the chemical alterations associated with neuronal death and demyelination [52]. Reduced levels of N-acetylaspartate may be associated with early brain injury and long-term outcomes in patients with TBI [53]. In mild TBI, 1H-MRS readings have been shown to vary even after concussion symptoms are no longer noticeable [54, 55]. Magnetic resonance elastography (MRE) is non-invasive way to measure mechanical function in the brain using MRI pulses to create acoustic wave propagation from which to measure tissue displacement [56]. This measurement of brain “stiffness” has been used to study several other clinical concerns, such as multiple sclerosis and aging, and may be useful in the prognosis of TBI [5].

Though it is not currently supported by the Brain Trauma Foundation guidelines, near-infrared spectroscopy (NIRS) is a promising non-invasive method for prehospital screening of intracranial bleeding. NIRS uses chromophore absorption to detect fluid and hemoglobin oxygenation near the brain, which can help to detect executive dysfunction in post-TBI patients with neurocognitive disorders. One study compared handheld NIRS Infrascanner scan time, ease-of-use, and change in treatment compared with CT in the pre-hospital screening of TBI. The Infrascanner had a sensitivity of 93.3% and a specificity of 78.6%, took from 1.5–10 minutes to perform, and had a median ease-of-use of 7 out of 10. On the downside, the Infrascanner detected three false positive and one false negative, is difficult to obtain scans of the dorsal occipital site when the patient is supine, did not significantly change the ultimate course of treatment of any patients, and emits an audio signal when scanning is complete, which can be difficult to hear in noisy environments [57].

The Brain Trauma Foundation also does not endorse the use of transcranial doppler (TCD) ultrasonography due to lack of evidence of its usefulness. Despite this, TCD is commonly used in the management of TBI; one study found it to be the second most used form of ICP monitoring at the patient’s bedside and that 40% of neuro-intensive care units use TCD in the management of TBI [58]. The use of TCD may be questioned due to the dependency on user-experience, but robotic TCDs are being investigated as an alternative that could eliminate this tendency for user error and allow TCD to become more reliable in monitoring TBI [5].

4. Automated Reading and Processing of Data

The sheer volume of imaging data requires clinical settings to adopt a system that can systematically and thoroughly analyze and predict the status of TBI patients. In addition to this, manual analyzation of imaging data can be time consuming and prone to human error, potentially leading to delayed intervention and misdiagnosis of cerebral injury. Convolutional neural networks (CNNs) utilize biological neural machine learning to interpret brain imaging by hierarchically breaking down and comparing complex images through pattern recognition. In the future, CNNs could be used to specify protocols to best treat brain injuries based on the specific neural networks interrupted [59]. One study showed that CNNs can detect microbleeds with a similar accuracy to experienced radiologists, though there are many false positives associated with automatic algorithms for detecting cerebral microbleeds. This study suggested a two-stage detection framework based on 3D fast radial symmetry transform of images from SWI and false positive reduction by CNN analysis of high-pass filtered phase images through CNN to improve the utility of automatic TBI imaging. The suggested two-stage cerebral microbleed detection algorithm had an optimal sensitivity of 95.8%, precision of 70.9%, and 1.6 false positives per case. This performance is comparable with experienced human raters and demonstrates the applicability of deep learning techniques to imaging analysis [60]. Another advancement in the processing of TBI imaging data is the use of automated methods of MRI segmentation to increase the speed and accuracy of MRI readings. Like the detection of cerebral microbleeds, the use of region segmentation is critical in the processing of MRI data and is often performed manually, which is time consuming and can result in user-error variability. There may be benefit in investigating how deep learning algorithms can be applied to automated MRI segmentation frameworks to improve the ease and quality of imaging analysis [61].

Nanoparticles are a recent advancement in TBI treatment that could increase the site-specific delivery of TBI therapy by promoting the accumulation and retention of these treatments to specific injured regions of the brain. Currently, nanoparticle delivery systems are either limited by single imaging modalities or have multimodal imaging capabilities, but are limited by complicated synthesis methods. One study suggested that mixed lanthanide oxide magnetic nanoparticles with a ultrasmall 2 nanometer core and hydrodynamic size of 13.5 nanometers can be detected by multimodality imaging using high spatial fluorescence imaging and temporal MRI frequencies and can quickly accumulate and be retained by brain parenchyma [62].

In addition to the advancements in automated imaging reading using CNNs and multimodality models, there are several preclinical TBI imaging strategies being developed to more accurately model TBI under experimental conditions. For example, some animal studies have shown that using fMRI to better characterize structural and functional changes in gray and white matter may provide a novel way to diagnose low grade TBI that cannot be efficiently classified with traditional CT and MRI scanning. When used alongside other imaging, these methods can provide data on diffusivity and fractional anisotropy in specific regions of the brain, as well as the degree of diffuse axonal injury, based on myelin and diminished microstructural integrity of the brain [63]. In 2014, one of the first animal studies to establish that interrupted networks and functional connectivity abnormalities between neural tissue injured during TBI and specific brain regions were associated with functional status post-TBI was conducted [64]. More recent animal studies have built upon this to demonstrate how fMRI and DTI can be used to track diffused and persistent neural damage post-TBI, based on neuronal connectivity, axonal integrity, and neurovascular function. fMRI-based resting-state functional connectivity (RSFC) was used in one study to measure neural connectivity and further showed that decreased RSFC strength in the cortex, hippocampus, and thalamus, as well as an increase in interhemispheric asymmetry, were more common in rat models with fluid percussion injury reminiscent of TBI [65]. Recent animal studies have also shown that using microarrays to detect long non-coding RNA (lncRNA) can be helpful in further understanding the pathophysiology of the TBI. Rats with induced TBI were shown to have alterations in messenger RNA (mRNA) and microRNA (miRNA) in their neural tissue, shortly following the injury. While these pieces of genetic material do not directly correspond to protein, they can induce the expression of molecules that may be involved in the progression of injury after the initial insult. One study found that the most common pathways that were aberrantly activated by lncRNA following surgically-induced, TBI-like injury in rats were inflammation and apoptosis, two of the major processes that contribute to secondary, long-term TBI morbidity. Energy metabolism, chemokine activation, hypoxia, and DNA transcription were also significantly altered in the experimental rats. Using animal models to study the genetic implications of TBI can provide a physiological rationale on which to base future imaging and treatment modalities [66].

5. Conclusions

This review provided an overview of some of the recent imaging improvements seen in the diagnosis and treatment of TBI. With the massive quantity of information available and the promise of even more knowledge from future advancements, it seems that one of the primary concerns moving forward is implementing multimodality imaging and developing reliable algorithms for the automated processing of this data through CNNs. It is also apparent that the use of certain imaging modalities such as NIRS and TCD is not standardized, and that consensus regarding acceptable ranges of certain parameters such as fractional anisotropy and mean diffusivity is lacking. Early TBI imaging can be an immensely powerful tool for acute treatment, but with recent literature revealing the potentially debilitating long-term effects of even low severity TBI, such as sports-associated concussions, advancements in TBI imaging could be an equally important tool for preventing chronic injuries that manifest long after the patient leaves the clinic.

Author Contributions

Conceptualization and overview by BLW. AA selected the relevant references and made substantial contributions to the conception and design of this manuscript. Both authors contributed to editorial changes in the manuscript. Both authors read and approved the final manuscript. Both authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest. Brandon Lucke-Wold is serving as one of guest editors of this journal. We declare that Brandon Lucke-Wold had no involvement in the peer review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to Gernot Riedel.

References
[1]
CDC. TBI Data. 2022. Available at: https://www.cdc.gov/traumaticbraininjury/data/index.html (Accessed: 21 March 2022).
[2]
Carney N, Totten AM, O’Reilly C, Ullman JS, Hawryluk GW, Bell MJ, et al. Guidelines for the Management of Severe Traumatic Brain Injury, Fourth Edition. Neurosurgery. 2017; 80: 6–15.
[3]
Hawryluk GWJ, Aguilera S, Buki A, Bulger E, Citerio G, Cooper DJ, et al. A management algorithm for patients with intracranial pressure monitoring: the Seattle International Severe Traumatic Brain Injury Consensus Conference (SIBICC). Intensive Care Medicine. 2019; 45: 1783–1794.
[4]
McCredie VA, Alali AS, Scales DC, Rubenfeld GD, Cuthbertson BH, Nathens AB. Impact of ICU Structure and Processes of Care on Outcomes After Severe Traumatic Brain Injury: A Multicenter Cohort Study. Critical Care Medicine. 2018; 46: 1139–1149.
[5]
Smith LGF, Milliron E, Ho ML, Hu HH, Rusin J, Leonard J, et al. Advanced neuroimaging in traumatic brain injury: an overview. Neurosurgical Focus. 2019; 47: E17.
[6]
Menon DK, Ercole A. Critical care management of traumatic brain injury. Handbook of Clinical Neurology. 2017; 140: 239–274.
[7]
Lindblad C, Raj R, Zeiler FA, Thelin EP. Current state of high-fidelity multimodal monitoring in traumatic brain injury. Acta Neurochirurgica. 2022; 164: 3091–3100.
[8]
Capizzi A, Woo J, Verduzco-Gutierrez M. Traumatic Brain Injury: An Overview of Epidemiology, Pathophysiology, and Medical Management. Medical Clinics of North America. 2020; 104: 213–238.
[9]
Teasdale G, Maas A, Lecky F, Manley G, Stocchetti N, Murray G. The Glasgow Coma Scale at 40 years: standing the test of time. The Lancet Neurology. 2014; 13: 844–854.
[10]
Bodien YG, Barra A, Temkin NR, Barber J, Foreman B, Vassar M, et al. Diagnosing Level of Consciousness: The Limits of the Glasgow Coma Scale Total Score. Journal of Neurotrauma. 2021; 38: 3295–3305.
[11]
Brown JB, Forsythe RM, Stassen NA, Peitzman AB, Billiar TR, Sperry JL, et al. Evidence-based improvement of the National Trauma Triage Protocol: The Glasgow Coma Scale versus Glasgow Coma Scale motor subscale. Journal of Trauma and Acute Care Surgery. 2014; 77: 95–102.
[12]
Shetty VS, Reis MN, Aulino JM, Berger KL, Broder J, Choudhri AF, et al. ACR Appropriateness Criteria Head Trauma. Journal of the American College of Radiology. 2016; 13: 668–679.
[13]
Schweitzer AD, Niogi SN, Whitlow CT, Tsiouris AJ. Traumatic Brain Injury: Imaging Patterns and Complications. Radiographics. 2019; 39: 1571–1595.
[14]
Jamjoom AAB, Rhodes J, Andrews PJD, Grant SGN. The synapse in traumatic brain injury. Brain. 2021; 144: 18–31.
[15]
Paterakis K, Karantanas AH, Komnos A, Volikas Z. Outcome of patients with diffuse axonal injury: the significance and prognostic value of MRI in the acute phase. Journal of Trauma and Acute Care Surgery. 2000; 49: 1071–1075.
[16]
Adams JH, Doyle D, Ford I, Gennarelli TA, Graham DI, McLellan DR. Diffuse axonal injury in head injury: definition, diagnosis and grading. Histopathology. 1989; 15: 49–59.
[17]
Skandsen T, Kvistad KA, Solheim O, Strand IH, Folvik M, Vik A. Prevalence and impact of diffuse axonal injury in patients with moderate and severe head injury: a cohort study of early magnetic resonance imaging findings and 1-year outcome. Journal of Neurosurgery. 2010; 113: 556–563.
[18]
Luccichenti G, Giugni E, Péran P, Cherubini A, Barba C, Bivona U, et al. 3 Tesla is twice as sensitive as 1.5 Tesla magnetic resonance imaging in the assessment of diffuse axonal injury in traumatic brain injury patients. Functional Neurology. 2010; 25: 109–114.
[19]
Studerus-Germann AM, Gautschi OP, Bontempi P, Thiran JP, Daducci A, Romascano D, et al. Central nervous system microbleeds in the acute phase are associated with structural integrity by DTI one year after mild traumatic brain injury: A longitudinal study. Neurologia i Neurochirurgia Polska. 2018; 52: 710–719.
[20]
Toth A, Kornyei B, Kovacs N, Rostas T, Buki A, Doczi T, et al. Both hemorrhagic and non-hemorrhagic traumatic MRI lesions are associated with the microstructural damage of the normal appearing white matter. Behavioural Brain Research. 2018; 340: 106–116.
[21]
van der Horn HJ, de Haan S, Spikman JM, de Groot JC, van der Naalt J. Clinical relevance of microhemorrhagic lesions in subacute mild traumatic brain injury. Brain Imaging and Behavior. 2018; 12: 912–916.
[22]
de Haan S, de Groot JC, Jacobs B, van der Naalt J. The association between microhaemorrhages and post - traumatic functional outcome in the chronic phase after mild traumatic brain injury. Neuroradiology. 2017; 59: 963–969.
[23]
Hütter BO, Altmeppen J, Kraff O, Maderwald S, Theysohn JM, Ringelstein A, et al. Higher sensitivity for traumatic cerebral microbleeds at 7 T ultra-high field MRI: is it clinically significant for the acute state of the patients and later quality of life? Therapeutic Advances in Neurological Disorders. 2020; 13: 1756286420911295.
[24]
Studerus-Germann AM, Thiran JP, Daducci A, Gautschi OP. Diagnostic approaches to predict persistent post-traumatic symptoms after mild traumatic brain injury - a literature review. International Journal of Neuroscience. 2016; 126: 289–298.
[25]
Mukherjee P, Berman JI, Chung SW, Hess CP, Henry RG. Diffusion tensor MR imaging and fiber tractography: theoretic underpinnings. American Journal of Neuroradiology. 2008; 29: 632–641.
[26]
Saatman KE, Duhaime AC, Bullock R, Maas AI, Valadka A, Manley GT. Workshop Scientific Team and Advisory Panel Members. Classification of traumatic brain injury for targeted therapies. Journal of Neurotrauma. 2008; 25: 719–738.
[27]
Bae CR, Na Y, Cho M, Hwang YM, Tae WS, Pyun SB. Structural Changes in the Arcuate Fasciculus and Recovery of Post-stroke Aphasia: A 6-Month Follow-up Study using Diffusion Tensor Imaging. Neurorehabilitation and Neural Repair. 2022; 36: 633–644.
[28]
Bazarian JJ, Zhong J, Blyth B, Zhu T, Kavcic V, Peterson D. Diffusion tensor imaging detects clinically important axonal damage after mild traumatic brain injury: a pilot study. Journal of Neurotrauma. 2007; 24: 1447–1459.
[29]
Yuh EL, Cooper SR, Mukherjee P, Yue JK, Lingsma HF, Gordon WA, et al. Diffusion tensor imaging for outcome prediction in mild traumatic brain injury: a TRACK-TBI study. Journal of Neurotrauma. 2014; 31: 1457–1477.
[30]
Sidaros A, Engberg AW, Sidaros K, Liptrot MG, Herning M, Petersen P, et al. Diffusion tensor imaging during recovery from severe traumatic brain injury and relation to clinical outcome: a longitudinal study. Brain. 2008; 131: 559–572.
[31]
Wilde EA, Ayoub KW, Bigler ED, Chu ZD, Hunter JV, Wu TC, et al. Diffusion tensor imaging in moderate-to-severe pediatric traumatic brain injury: changes within an 18 month post-injury interval. Brain Imaging and Behavior. 2012; 6: 404–416.
[32]
Wallace EJ, Mathias JL, Ward L. Diffusion tensor imaging changes following mild, moderate and severe adult traumatic brain injury: a meta-analysis. Brain Imaging and Behavior. 2018; 12: 1607–1621.
[33]
Brown SSG, Dams-O’Connor K, Watson E, Balchandani P, Feldman RE. Case Report: An MRI Traumatic Brain Injury Longitudinal Case Study at 7 Tesla: Pre- and Post-injury Structural Network and Volumetric Reorganization and Recovery. Frontiers in Neurology. 2021; 12: 631330.
[34]
Mokri B. The Monro-Kellie hypothesis: applications in CSF volume depletion. Neurology. 2001; 56: 1746–1748.
[35]
Grände PO. Critical Evaluation of the Lund Concept for Treatment of Severe Traumatic Head Injury, 25 Years after Its Introduction. Frontiers in Neurology. 2017; 8: 315.
[36]
Doshi H, Wiseman N, Liu J, Wang W, Welch RD, O’Neil BJ, et al: Cerebral hemodynamic changes of mild traumatic brain injury at the acute stage. PLoS ONE. 2015; 10: e0118061.
[37]
Kim J, Whyte J, Patel S, Avants B, Europa E, Wang J, et al. Resting cerebral blood flow alterations in chronic traumatic brain injury: an arterial spin labeling perfusion FMRI study. Journal of Neurotrauma. 2010; 27: 1399–1411.
[38]
Douglas DB, Chaudhari R, Zhao JM, Gullo J, Kirkland J, Douglas PK, et al. Perfusion Imaging in Acute Traumatic Brain Injury. Neuroimaging Clinics of North America. 2018; 28: 55–65.
[39]
Soustiel JF, Mahamid E, Goldsher D, Zaaroor M. Perfusion-CT for early assessment of traumatic cerebral contusions. Neuroradiology. 2008; 50: 189–196.
[40]
Metting Z, Rödiger LA, Stewart RE, Oudkerk M, De Keyser J, van der Naalt J. Perfusion computed tomography in the acute phase of mild head injury: regional dysfunction and prognostic value. Annals of Neurology. 2009; 66: 809–816.
[41]
Bendinelli C, Cooper S, Evans T, Bivard A, Pacey D, Parson M, et al. Perfusion Abnormalities are Frequently Detected by Early CT Perfusion and Predict Unfavourable Outcome Following Severe Traumatic Brain Injury. World Journal of Surgery. 2017; 41: 2512–2520.
[42]
Tavakoli S, Peitz G, Ares W, Hafeez S, Grandhi R. Complications of invasive intracranial pressure monitoring devices in neurocritical care. Neurosurgical Focus. 2017; 43: E6.
[43]
Volovici V, Pisică D, Gravesteijn BY, Dirven CMF, Steyerberg EW, Ercole A, et al. Comparative effectiveness of intracranial hypertension management guided by ventricular versus intraparenchymal pressure monitoring: a CENTER-TBI study. Acta Neurochirurgica. 2022; 164: 1693–1705.
[44]
Thelin EP, Nelson DW, Ghatan PH, Bellander BM. Microdialysis Monitoring of CSF Parameters in Severe Traumatic Brain Injury Patients: A Novel Approach. Frontiers in Neurology. 2014; 5: 159.
[45]
Zeiler FA, Thelin EP, Helmy A, Czosnyka M, Hutchinson PJA, Menon DK. A systematic review of cerebral microdialysis and outcomes in TBI: relationships to patient functional outcome, neurophysiologic measures, and tissue outcome. Acta Neurochirurgica. 2017; 159: 2245–2273.
[46]
Guilfoyle MR, Helmy A, Donnelly J, Stovell MG, Timofeev I, Pickard JD, et al. Characterising the dynamics of cerebral metabolic dysfunction following traumatic brain injury: A microdialysis study in 619 patients. PLoS ONE. 2021; 16: e0260291.
[47]
Timofeev I, Carpenter KL, Nortje J, Al-Rawi PG, O’Connell MT, Czosnyka M, et al. Cerebral extracellular chemistry and outcome following traumatic brain injury: a microdialysis study of 223 patients. Brain. 2011; 134: 484–494.
[48]
Jilka SR, Scott G, Ham T, Pickering A, Bonnelle V, Braga RM, et al. Damage to the Salience Network and interactions with the Default Mode Network. Journal of Neuroscience. 2014; 34: 10798–10807.
[49]
Raichle ME, MacLeod AM, Snyder AZ, Powers WJ, Gusnard DA, Shulman GL. A default mode of brain function. Proceedings of the National Academy of Sciences of the United States of America. 2001; 98: 676–682.
[50]
Laureys S, Celesia GG, Cohadon F, Lavrijsen J, León-Carrión J, Sannita WG, et al. Unresponsive wakefulness syndrome: a new name for the vegetative state or apallic syndrome. BMC Medicine. 2010; 8: 68.
[51]
Di Perri C, Bahri MA, Amico E, Thibaut A, Heine L, Antonopoulos G, et al. Neural correlates of consciousness in patients who have emerged from a minimally conscious state: a cross-sectional multimodal imaging study. The Lancet Neurology. 2016; 15: 830–842.
[52]
de Figueiredo EH, Borgonovi AF, Doring TM. Basic concepts of MR imaging, diffusion MR imaging, and diffusion tensor imaging. Magnetic Resonance Imaging Clinics. 2011; 19: 1–22.
[53]
Holshouser B, Pivonka-Jones J, Nichols JG, Oyoyo U, Tong K, Ghosh N, et al. Longitudinal Metabolite Changes after Traumatic Brain Injury: A Prospective Pediatric Magnetic Resonance Spectroscopic Imaging Study. Journal of Neurotrauma. 2019; 36: 1352–1360.
[54]
Giacino JT, Katz DI, Schiff ND, Whyte J, Ashman EJ, Ashwal S, et al. Comprehensive systematic review update 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. 2018; 91: 461–470.
[55]
Gardner A, Iverson GL, Stanwell P. A systematic review of proton magnetic resonance spectroscopy findings in sport-related concussion. Journal of Neurotrauma. 2014; 31: 1–18.
[56]
Muthupillai R, Ehman RL. Magnetic resonance elastography. Nature Medicine. 1996; 2: 601–603.
[57]
Peters J, Van Wageningen B, Hoogerwerf N, Tan E. Near-Infrared Spectroscopy: A Promising Prehospital Tool for Management of Traumatic Brain Injury. Prehospital and Disaster Medicine. 2017; 32: 414–418.
[58]
Gomez A, Batson C, Froese L, Sainbhi AS, Zeiler FA. Utility of Transcranial Doppler in Moderate and Severe Traumatic Brain Injury: A Narrative Review of Cerebral Physiologic Metrics. Journal of Neurotrauma. 2021; 38: 2206–2220.
[59]
Chartrand G, Cheng PM, Vorontsov E, Drozdzal M, Turcotte S, Pal CJ, et al. Deep Learning: A Primer for Radiologists. Radiographics. 2017; 37: 2113–2131.
[60]
Liu S, Utriainen D, Chai C, Chen Y, Wang L, Sethi SK, et al. Cerebral microbleed detection using Susceptibility Weighted Imaging and deep learning. NeuroImage. 2019; 198: 271–282.
[61]
De Feo R, Hämäläinen E, Manninen E, Immonen R, Valverde JM, Ndode-Ekane XE, et al. Convolutional Neural Networks Enable Robust Automatic Segmentation of the Rat Hippocampus in MRI After Traumatic Brain Injury. Frontiers in Neurology. 2022; 13: 820267.
[62]
Bony BA, Miller HA, Tarudji AW, Gee CC, Sarella A, Nichols MG, et al. Ultrasmall Mixed Eu-Gd Oxide Nanoparticles for Multimodal Fluorescence and Magnetic Resonance Imaging of Passive Accumulation and Retention in TBI. ACS Omega. 2020; 5: 16220–16227.
[63]
Sinke MRT, Otte WM, Meerwaldt AE, Franx BAA, Ali MHM, Rakib F, et al. Imaging Markers for the Characterization of Gray and White Matter Changes from Acute to Chronic Stages after Experimental Traumatic Brain Injury. Journal of Neurotrauma. 2021; 38: 1642–1653.
[64]
Mishra AM, Bai X, Sanganahalli BG, Waxman SG, Shatillo O, Grohn O, et al. Decreased resting functional connectivity after traumatic brain injury in the rat. PLoS ONE. 2014; 9: e95280.
[65]
Parent M, Li Y, Santhakumar V, Hyder F, Sanganahalli BG, Kannurpatti SS. Alterations of Parenchymal Microstructure, Neuronal Connectivity, and Cerebrovascular Resistance at Adolescence after Mild-to-Moderate Traumatic Brain Injury in Early Development. Journal of Neurotrauma. 2019; 36: 601–608.
[66]
Wang CF, Zhao CC, Weng WJ, Lei J, Lin Y, Mao Q, et al. Alteration in Long Non-Coding RNA Expression after Traumatic Brain Injury in Rats. Journal of Neurotrauma. 2017; 34: 2100–2108.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top