Open access peer-reviewed chapter

Recent Advances in the Development of Biofluid-Based Prognostic Biomarkers of Diffuse Axonal Injury

Written By

Vinu V. Gopal, Rinku Raj Mullasseril and Goutam Chandra

Submitted: 09 April 2022 Reviewed: 14 April 2022 Published: 03 June 2022

DOI: 10.5772/intechopen.104933

From the Edited Volume

Frontiers In Traumatic Brain Injury

Edited by Xianli Lv, Yi Guo and Gengsheng Mao

Chapter metrics overview

708 Chapter Downloads

View Full Metrics

Abstract

Even though head injury is a silent pandemic of the century producing immense social and economic impact, predictive models have not been established to develop strategies promoting the development of reliable diagnostic tools and effective therapeutics capable of improving the prognosis. Diffuse axonal injury (DAI) is a type of traumatic brain injury (TBI) that results from a blunt injury to the brain. Discovering biomarkers for DAI have been a matter of debate and research. A number of studies have reported biomarkers that are correlated with severity of TBI but no conclusive and reproducible clinical evidence regarding the same has been put forward till now. Additionally, many DAI biomarkers have limitations so that they cannot be generalized for universal applications. The properties of these biomarkers should be extensively researched along with the development of novel biomarkers to aid important clinical decisions for the benefit of the society. This chapter summarizes the existing biofluid-based biomarkers, critically examines their limitations and highlights the possibilities of a few novel biomolecules as prognostic biomarkers of DAI.

Keywords

  • diffuse axonal injury
  • biofluid
  • neuronal damage
  • prognosis
  • rehabilitation

1. Introduction

Central nervous system (CNS) trauma including traumatic brain injury (TBI) is a major cause of long-term injury, disability and death among young adults worldwide [1, 2]. Around 1.6 million individuals suffer from TBI every year in India with 200,000 deaths [3]. In the USA, there were more than 223,000 TBI-related hospitalizations in 2018 and about 166 Americans died from TBI-related injury each day in 2019 [4]. These estimates do not include the many TBIs that are only treated in the emergency department, primary care, urgent care, or those that go untreated [5]. Since the affected individuals are disabled and out of work during the most productive period of their life, these devastating conditions have an enormous physical, mental and economic burden to the country.

TBI is a heterogeneous neurological condition, ranging from single or repetitive concussion /mild TBI to penetrating head injury, focal contusion, different forms of hematoma (subdural and epidural) and diffuse injury. Depending on the motor, verbal, and eye-opening responses of the affected individuals, the Glasgow coma scale (GCS) was designed to access the disability. The GCS measures the following three functions: Motor response (scores: 6-normal, 5-localized to pain, 4-withdraws to pain, 3-flexion response to pain, 2-extension response to pain, 1-no motor response), Verbal response (scores: 5-normal conversation, 4-oriented conversation, 3-words, but not coherent, 2-no words, only sounds, 1-none), and Eye-opening response (scores: 4-spontaneous, 3-to voice, 2-to pain, 1-none). Based on the GCS score, TBI is classified as mild, moderate, or severe. TBI patients with GCS of 13 to 15 are classified to be mild, which includes the majority of these patients. Patients with a GCS of 9 to 12 are considered to have a moderate TBI, while patients with a GCS below eight are classified as having a severe TBI [6].

The heterogeneous nature of TBI with respect to severity of the injury and comorbidities make patient outcome difficult to predict. While mild TBI or concussion may affect neural cells temporarily, severe TBI is associated with substantial axonal injury and physical damage to the brain, which can result in blood-brain barrier disruption and neuroinflammatory changes [7]. Moderate to severe TBI can usually be visible as structural abnormalities using radiological examinations such as computed tomography (CT) or magnetic resonance imaging (MRI). However, more subtle neural alterations characteristic of mild TBI are not easily detected by these imaging techniques.

Although TBI is an extremely complex condition, there have been many advances in recent years in relation to the diagnosis, monitoring and treatment of the affected patients. Shortcomings in our knowledge of the physiopathology of TBI and the development of reliable predictive models capable of offering an early orientation as to the patient outcome, will improve the diagnostic and therapeutic strategies on an individualized basis. Likewise, we need valid predictive models in severe TBI in order to define efficacy endpoints in the evaluation of new drugs or treatment strategies–since the usual primary endpoints (death and disability) are widely recognized as being inadequate and could explain the discouraging results obtained with certain promising drugs.

Advertisement

2. Pathogenesis of DAI

Diffuse axonal injury (DAI) is a type of TBI that results from a blunt injury to the brain, which happens when the brain rapidly shifts inside the skull as an injury is occurring. It is one of the most common but devastating types of TBI. Neuronal injuries associated with DAI fall into two categories: (i) primary injury, which is directly caused by mechanical forces during the initial insult; and (ii) secondary injury, which is caused as a consequence of primary injury to further tissue and cellular damages.

2.1 Primary brain injuries

Primary brain injuries refer to the sudden and profound injury to the brain that occurs at the time of the motor vehicle accident, gunshot wound, or accidental fall. The immediate impact of different mechanical insults to the brain can cause two types of primary TBI: focal and diffuse brain injuries. Focal TBI generally results from a blow to the head that produces cerebral contusions or hematomas. Epidural hematomas, subdural hematomas, and cerebral contusions are the results of focal brain injuries [8]. In contrast, diffuse lesions (also known as DAI) are seen more commonly in lesions that involve rapid acceleration, deceleration, or rotational forces. DAI accounts for about 70% of TBI cases. The sites that are most prone to DAI are the reticular formation, basal ganglia, superior cerebellar peduncles, limbic fornices, hypothalamus and corpus callosum [8]. Interestingly, both types of injuries may co-exist in patients who suffered from moderate to severe TBI [9].

Brain injuries may occur in one of two ways: closed brain TBI and penetrating TBI. Closed brain TBIs occur when there is a non-penetrating injury to the brain with no break in the skull. A closed brain injury is caused by a rapid forward or backward movement and shaking of the brain inside the bony skull that result in bruising and tearing of brain tissue and blood vessels. In contrast, Penetrating, or open head injuries occur when there is a break in the skull, caused by hitting with a sharp object such as a bullet. These injuries exhibit focal brain damage due to lacerations, compression and concussion forces with evidence of skull fracture and localized contusion (Figure 1) at the core of injury site, known as the ‘coup’ area [10, 11]. Compromised blood supply at the coup area due to epidural, subdural and intracerebral hemorrhages and hematomas might result in necrosis of neuronal and glial cells at confined layers of the brain. Secondary contusion may develop in brain tissues opposite to or surrounding the coup area due to secondary impact when the brain rebounds and strikes the skull [11].

Figure 1.

Schematic representation of the pathogenesis of TBI. TBI may be divided into primary injury and secondary injury. Primary TBI results from mechanical injury at the time of insult, while secondary injury is caused by the physiologic responses to the initial injury. Primary brain injury comprises the direct physical injury to the brain such as compression, deformation, displacement, stretching, shearing, tearing, and crushing of brain which results in damage to vasculature, neural, and glial tissues. Most of the neurological damage from TBI is due to the secondary injury which evolves over the ensuing hours and days after the initial injury or impact. The mechanisms by which TBI trigger neurodegeneration are areas of active research. Previous investigations found roles of excitotoxicity, loss of cerebral autoregulation, blood-brain barrier compromise, mitochondrial dysfunction, oxidative stress, lipid peroxidation, neuroinflammation, axon degeneration, impaired autophagy and apoptotic cell death in the development of neurodegeneration following brain injury. TBI produces both acute and chronic consequences that lead to permanent disabilities that increase long-term mortality and reduced life expectancy. The direct consequences of a single or repetitive TBI can result in various secondary pathological conditions, including seizures, sleep disorders, neurodegenerative diseases, neuroendocrine dysregulation, and psychiatric problems. Changes initiated by TBI can persist for weeks to months or even years after injury and significantly affect quality-of-life of the affected victims.

2.2 Secondary brain injuries

Secondary brain injuries refer to the changes that evolve over a period of time after the primary brain injury. The biochemical, cellular and physiological events that occur during primary injury often progress into delayed and prolonged secondary damaging cascade of cellular, chemical, tissue, or blood vessel changes in the brain that contribute to further destruction of brain tissue. Secondary brain injuries can last from hours to days and even weeks and may be caused by impairment or local declines in cerebral blood flow resulting in local edema, hemorrhage, or increased intracranial pressure and even brain herniation. Other types of secondary injury due to TBI include hypercapnia, acidosis, meningitis and brain abscess [12]. Mechanistically, a number of factors contribute to these changes, which include excitotoxicity, loss of cerebral autoregulation, blood-brain barrier compromise, mitochondrial dysfunction, oxidative stress, lipid peroxidation, neuroinflammation, axon degeneration, impaired autophagy and apoptotic cell death (Figure 1) [10, 13].

The hallmark feature of DAI is extensive damage of axons predominantly in subcortical and deep white matter tissue, which leads to impairment of axonal transport and degradation of axonal cytoskeleton. The strong tensile forces generated during primary injury by rapid deceleration and acceleration of the brain due to multiple non-contact forces causing shearing and stretching injury in cerebral brain tissues damage neuronal axons, oligodendrocytes and blood vasculature, leading to brain edema and ischemic brain damage [14]. These axonal damages can persist for months after DAI.

The degree of axonal injury and neuronal degeneration determines the severity of TBI. While explosive blast TBI is a result of shock waves instead of inertial forces, it displays the characteristics of a typical DAI. Depending on the severity of the injury, patients may later develop cognitive deficits, behavioral changes and hemiparesis (Figure 1).

Advertisement

3. Need for biofluid-based brain damage biomarkers

While mild TBI or concussion may affect neural cells temporarily, severe TBI is associated with substantial axonal injury and physical damage to the brain, which can result in blood-brain barrier disruption and neuroinflammatory changes [7]. Although, moderate to extensive brain injury may be visible as structural abnormalities using CT scan or MRI, more subtle neural alterations characteristic of mild TBI are not easily detected by these imaging techniques. Moreover, changes due to DAI in the brain are often microscopic and may not be visualized on CT scan or MRI scans.

Mild TBI is highly prevalent in military populations, with many service members suffering from long-term symptoms [15]. It is also very common among road accident victims. The condition results from etiologies of neural contusion and axonal injury, which subsequently results in biochemical, metabolic, and cellular changes that may be responsible for some of the long-term problems observed in patients who develop post-concussion syndrome [16]. Moreover, moderate to severe TBI remains another important public health problem, due to the large percentage of unfavorable outcomes involved such as death and disabling sequelae. The huge treatment costs, associated compensations, disability pensions and years of income from work lost in affected individuals are the major financially devastating turns in the affected families. Therefore, identifying critical markers of neural injury in biofluids of these patients would be crucial for predicting long-term functional outcome and for taking rehabilitation decisions. Encouragingly, significant scientific advances on the TBI biomarker research in the last decade have increased our understanding of the complex and heterogeneous pathophysiological processes associated with this condition. Emerging evidence from multiple research teams suggests that biofluid-based TBI biomarkers may have the potential to diagnose the presence of TBI of different severities, and to predict outcome.

Advertisement

4. Biomarkers for TBI/DAI

A biomarker is defined as a quantifiable biological indicator specific for a given physiological or pathological condition. Based on clinical utility, biomarkers may be categorized as: 1) diagnostic biomarkers, which identify the presence or absence of TBI, 2) prognostic biomarkers, which inform the clinicians about expected outcomes in injured individuals, and 3) predictive biomarkers, which predict response to a specific intervention and can be used to monitor response to therapy. Identification of biological markers of TBI could offer a more precise indication of the extent and severity of TBI, independently of the prior biological substrate and of other circumstances that accompany severe TBI–thereby contributing to homogeneously define different patient categories and risk stratify the head injury. These can also serve to screen and identify patients who may expect an altered or complicated recovery or might develop neurobehavioral deficits during the latter part of their life. Such markers would facilitate

  • individualization of the intensity of TBI/DAI

  • improve knowledge of the physiopathology of brain damage

  • afford essential complementary information for the diagnosis

  • predict the outcome of these patients

  • timing of patient management

  • development of strategies for preventing or minimizing secondary damage

  • evaluation of neuroprotective effects of novel biomolecules.

4.1 Which is an ideal biomarker?

An ideal TBI/DAI biomarker should have

  • high specificity and sensitivity for the brain tissue

  • rapid appearance in accessible biofluids such as cerebrospinal fluid (CSF), plasma and/or whole blood immediately after irreversible brain tissue damage.

  • must be elevated in various forms and/or severities of brain damage in the acute phase (3 h to 24 h post-injury).

  • must reflect the extent and severity of the damage, as defined by GCS, CT abnormality, in due course of time.

  • variations between age and gender groups must be minimal.

  • must have low basal biofluid levels in non-injured healthy control population

  • should be responsive to therapeutic treatments

  • the tools for analysis and immediate detection of the marker must be available and reproducible.

  • determination of the marker must be clinically relevant.

The literature given below is a concise description of the principal investigational brain damage biomarkers with a description of the tissues in which they originate, the compartment from which samples were collected, their pathological serum concentrations, and the main prognostic features (Table 1).

BiomarkerLocalizationRole in prognosisDrawbacksReferences
Tau/c-tauNeuronal axons and astrocytesMay predict outcomes after severe TBINot fully characterized[17, 18, 19]
Extracellular spaceAltered levels in CSFContradictory findings[20, 21, 22]
MBPAxonal myelin sheath, oligodendroglial cellsElevated levels in serum and predict severity and outcomeLimited sensitivity[23, 24]
CK-BBastrocytesMay predict outcomes after severe TBILow sensitivity and specificity in polytrauma[25, 26, 27]
NSENeuronElevated serum levels and specific to neuronal tissue than glial cellLong half-life and are expressed in red blood cells[28, 29, 30, 31, 32]
S100BAstrocytesPredict severity and act as adjuvant markerReduced specificity; not suitable for children under 2 years[33, 34]
GFAPAstrocytesMay predict neurological outcomesAlso expressed by the Leydig cells of the testes[35, 36, 37]
UCH-L1Neuronal cell bodyIncreased serum levels in brain damageNot fully characterized[38, 39, 40]
SBDPsAxons and presynaptic neuronal endingsMay predict neurological outcomesNot brain specific and difficult to quantify[41, 42, 43, 44]
NfLNeuronal cytoplasmReleased in response to CNS neuronal damageReleased in response to neurodegeneration and neuroinflammation[45]

Table 1.

Current and emerging DAI biomarkers.

4.2 Tau protein

Tau protein is an axonal cytoskeleton-stabilizing protein (Figure 2) of molecular weight of 30–50 kDa that provides structural elements of the cytoskeleton that are crucial for neuronal protein flow [46, 47]. There are 6 different tau protein isomers, which is phosphorylated at many sites by kinases such as casein kinase II, tau tubulin kinases, glycogen synthase kinase 3β, and cyclin dependent kinase 5 [48, 49, 50]. While these are present in a stable, unfolded and monomeric morphology in a healthy brain, tau proteins exist in hyperphosphorylated state in several neurodegenerative diseases including Alzheimer’s disease (AD) [46, 51]. Interestingly, TBI has been indicated as a risk factor for later development of AD and other neurodegenerative conditions [52, 53, 54, 55, 56] .

Figure 2.

Origin of biomarkers of DAI. In normal brain, NSE and UCH-L1 are localized in neuronal cytoplasm, while tau/c-tau are restricted mainly to axons and its terminals. Although tau is abundant in the neurons of the CNS, astrocytes express very low levels of this protein and it can be secreted into the brain interstitial fluid. APP is a type I transmembrane protein expressed in many cell types, including neurons. Aβ is derived from APP by enzymatic cleavage and is released to extracellular space. MBP is a constituent of neuronal myelin sheath, which is produced by oligodendrocytes. Spectrins (precursors of SBDPs), specifically βIV-spectrin is concentrated at axon initial segments and nodes of Ranvier. GFAP, CK-BB and S100B are normally present in astrocytes. DAI not only injures pre- and post-synaptic neurons but also damages their synapses, axons, myelin sheaths and neighboring astrocytes, oligodendrocytes, blood microvasculature and even the extracellular matrix network. Damages to specific cells and cellular components during DAI enable release of various molecules contained in those cells into the extracellular space. The released molecules, including those present in extracellular space like Aβ enter the damaged blood vessels and may be detected in the circulation.

Physical trauma causes activation of a number of proteases, which cause release of tau protein fragments in cleaved tau (c-tau) into the blood and CSF [57, 58]. Studies showed that the c-tau levels in CSF increase in the first 24 h after severe TBI [17, 59, 60]. Plasma phosphorylated tau (p-tau) and p-tau/t-tau ratios have been demonstrated to distinguish patients with acute and chronic TBI from healthy controls [18]. Smith and colleagues (1999) have shown deposition of p-tau following TBI [61]. C-tau in CSF is shown to be a predictor of clinical outcomes in severe TBI subjects [60, 62]. Moreover, elevated c-tau could be a chronic manifestation in DAI, since tau is localized to the axons [59]. However, the practical role of this molecule in DAI has not been fully established.

4.3 Amyloid-β (Aβ) protein

Amyloid-β (Aβ) is a 4 kDa extracellular protein derived from amyloid precursor protein (APP), which is cleaved by secretase enzymes [63]. APP is a membrane protein expressed in both CNS (APP 695) and peripheral organs and tissues (APP 751 and APP 770) [64]. APP with 695 amino acids is present as glycosylated receptors on cell surface and is hydrolysed by α-secretase followed by γ-secretase under normal conditions to produce soluble Aβ through non-amyloidogenic pathway [63, 65]. In amyloidogenic pathway, the mutations in APP and components of α-secretase, presinillin 1 (PSEN1) and presinillin 2 (PSEN2) leads to the cleavage by β amyloid cleaving enzyme-1 (BACE1) and γ-secretase to form insoluble Aβ, Aβ1–40 and Aβ1–42. This amyloidogenic cleavage leads to extracellular accumulation of Aβ plaques, a pathological hallmark in AD [20, 65, 66]. APP plays an important role in cell adhesion processes and thus high concentrations are found at neuronal synaptic junction. Certain type of caspase breaks it down into a series of products which accumulate in cell bodies and axons. There are discordant evidence on the use of this protein as biomarker of TBI. One study of 29 patients with severe TBI revealed low levels of this protein in CSF probably due to reabsorption of the protein in the form of amyloid plaques [21]. As a contradiction to the above, Emmerling et al. (2020) found increasing levels in CSF after trauma and suggested that this could be a result of secondary axonal damage or loss of integrity of the BBB [22]. These contradictory findings, have caused Aβ-protein to be regarded as a non-reproducible biomarker and its potential role remains unclear requiring further research.

4.4 Myelin basic protein (MBP)

MBP (molecular weight of 18.5 kDa), found in oligodendroglial cells, is a key structural component of the multi-layered myelin sheath covering nerve fibers. The myelin sheath on neuronal axons serves as an insulator to increase the velocity of axonal impulse conduction. Due to the extended length of axonal fiber tracks, axons are particularly vulnerable to physical trauma to the brain. Thus, axonal injury is a common occurrence in both focal as well as diffuse brain trauma and can be found in TBI of all severities [14, 67]. As MBP maintains myelin structure by interacting with the lipids in the myelin membrane [68], axonal injury causes breakdown of the myelin sheath and release of MBP. This myelin-specific protein is also released into the bloodstream in cases of demyelinating diseases such as multiple sclerosis, or degradation by proteases, such as calpain [69, 70].

MBP is found to be elevated in serum after severe TBI in children [23, 24] and after mild TBI in adults [24]. Even though, it takes around 1–2 days to appear in the serum after TBI, the peak levels of MBP can persist for up to 2 weeks and can be a specific indicator for future intracranial hemorrhage [71]. However, as per present literature there is contradicting evidence for its role in TBI/DAI [72, 73, 74]. There was no difference in initial levels of serum MBP in a pediatric population with mild TBI when compared with controls, but there was a significant difference in the peak MBP levels between patients and controls [72]. MBP is also expressed on the myelin of peripheral nerves and its transcripts are present in the bone marrow and immune system and therefore it is not specific to the CNS. Even though serum levels are correlated with patient severity and outcome [71, 75] it has limited sensitivity as a marker for predicting severity of TBI [76].

4.5 Cerebral creatine kinase (CK) isoenzyme

CK isoenzymes are of three types: CK-1 (also known as CK-BB) is predominantly expressed in brain, lung, thyroid and prostate glands, gastrointestinal tract, urinary bladder, uterus and placenta. CK-2 (CK-MB) and CK-3 (CK-MM) are expressed in cardiac and skeletal muscles [77]. Brain tissue-specific CK-1 (CK-BB), with a molecular weight of 40–53 kDa, is found in astrocytes [25, 26]. A peak in serum cerebral CK concentration is observed in the first few hours after severe TBI and then gradually decrease and the marker remains high for days [78, 79, 80, 81]. In polytrauma, it remains persistently high without an initial dip [82, 83] . Its levels have been shown to rise significantly in CSF following hypoxic brain injury in cardiac arrest, which suggests that CK-BB release may be secondary to cerebral hypoperfusion due to systemic trauma [84]. The major limiting factor is that it has low sensitivity and specificity especially in cases of polytrauma [27, 85].

4.6 Neuron-specific enolase (NSE)

NSE, also known as γ -enolase or enolase 2, is a glycolytic enzyme with a molecular weight of 78 kDa and a half-life of 48 h. It exists as a homodimer (γ–γ) in mature neurons and neuroendocrine cells. The normal concentration of NSE in blood is <10 ng/ml. NSE elevations in blood compartment has been documented in severe as well as mild TBI [28, 29, 30, 31, 86]. Experimental models of trauma have correlated serum NSE to the severity of damage in TBI [87]. Major limitation of using NSE as specific biomarker for TBI is that it is also abundantly expressed in red blood cells [88]. Moreover, increased levels of NSE was also recorded previously in the serum of patients following non-traumatic brain damage such as ischemic events, intracerebral hemorrhage, cardiopulmonary resuscitation, secondary cerebral hypoxia etc. [89]. Some studies had correlated the biomarker to the development of DAI, though its behavior has not been clearly established in prospective trials.

NSE was initially suggested to be a very promising TBI severity marker due to its specificity to neuronal tissue than of glial cells. However, the results published till date has been contradictory on its role in predicting prognosis of patients with severe TBI. Long half-life is a major limiting factor for its use in trauma setting. Also, extracranial origin of NSE was demonstrated in hemorrhagic shock, long bone fracture, hemolysis, heart surgery, ischemia–reperfusion injury and malignant lung tumors making it a poor marker for TBI [90, 91, 92, 93, 94].

4.7 Glial fibrillary acidic protein (GFAP)

GFAP is a monomeric intermediate filament protein (molecular weight 52 kDa), present in the cytoskeleton of astrocytes in the brain. An increase in blood level of this biomarker suggests injury to the astrocytes and the BBB. Plasma concentrations >0.033 μg/l are regarded as pathological. Missler et al. (2002) were the first to propose the possible use of GFAP as an identifier of brain damage in serial serum measurements [95]. Later studies also confirmed that the serum concentration of this protein is not affected by extracranial injuries thus making it an effective biomarker for predicting poor outcome in the acute phase of severe TBI as well as for advocating the need for urgent neurosurgical procedures [41, 96]. GFAP (52 kDa) or its breakdown products (44–38 kDa) are released from injured brain tissue into biofluids such as CSF and enter the bloodstream after crossing the BBB with an early plasma peak (within 3 to 34 h) following brain injury [97, 98]. The blood levels then decrease gradually over the first week, starting from third day of injury.

Previous studies demonstrated that GFAP levels show an unfavorable outcome in patients with moderate or severe TBI [41, 96, 99, 100]. However, this may not be the case in patients with mild TBI due to the contamination from other sources [101, 102, 103]. However, Serum GFAP levels were also significantly higher in patients who died or had an unfavorable outcome [104]. Moreover, GFAP levels have correctly predicted neurological outcome at 6 months [35, 36, 104]. Furthermore, serum GFAP measured on day 1 of injury in pediatric TBI cases significantly correlated with functional outcomes at 6 months [105]. Thus, GFAP can be considered as an ideal biomarker of brain damage when combined with clinical variables though multicenter studies are needed for further validation.

4.8 S100-calcium binding protein B (S100B)

S100B, the most widely studied brain damage biomarker, is a low molecular weight (11 kDa) calcium binding protein of astroglial origin [33]. The homodimeric beta-subtype of S100 proteins (S100B) is synthesized in astrocytes of the CNS and in Schwann cells of the peripheral nerves, where it regulates intracellular calcium levels [106, 107, 108]. S100B localizes to the nucleus and cytoplasm associating with endomembranes, the centrosomes, microtubules and type III intermediate filaments [109]. The protein is naturally secreted by astrocytes into the extracellular space. Low amounts of S100B can cross the BBB and enter the microvasculature. Elevated levels of S100B in the serum were observed in TBI patients as well as in patients suffering from neurodegenerative diseases [110]. The serum levels of the protein have been associated with clinical severity, radiological severity, and an unfavorable outcome [111, 112, 113, 114] .

The biological function of this protein has not been fully established till date, though it is known to participate in neurogenesis, astrocytosis and axonal elongation. However, the molecule can also be produced and found outside the CNS, e.g., in kidney epithelial cells, ependymocytes, chondrocytes, adipocytes, melanocytes, Langerhans cells, dendritic cells, certain lymphocyte subpopulations, skeletal myofibers, myoblasts and muscle satellite cells [109]. Metabolism takes place in the kidneys, followed by excretion in urine, with an approximate half-life of 30–113 min, and is not affected by hemolytic phenomena [115]. Its role in urine level also needs further validated study. Since S100B can also be released from adipose tissue and cardiac/skeletal muscles, its levels are also elevated in orthopedic trauma without head injury [116]. Despite these confounders, S100B is actually a sensitive TBI biomarker for predicting CT abnormality and post-concussive syndrome development [117, 118, 119]. A number of previous studies have shown that S100B can actually differentiate between mild and severe TBI [120, 121].

The maximum serum concentration of S100B is reached 20 min after brain damage. The normal upper limit for this protein in relation to the detection of intracranial damage was defined as 0.1 μg/l based on a multicenter study in patients with mild TBI [122]. The measurement of S100B-protein can be influenced by patient age and gender in CSF samples but not in serum samples thus making it a practically feasible biomarker.

Some studies have determined its usefulness as a predictor of mortality, establishing orientative serum cut-off points for predicting a course leading to death or an unfavorable outcome [123, 124, 125]. On the other hand, S100B level has been correlated to the presence of secondary lesions, the extent of diffuse brain damage, and to modifications in intracranial pressure following different release patterns [126, 127]. S100B levels can also detect brain death development or mortality after severe TBI [128, 129]. Interestingly, serum levels of S100B > 0.7 ng/mL were reported to correlate with 100% mortality [130].

Another possible application of this protein refers to its time course according to the severity of the patient condition. A number of studies have documented persistently elevated serum levels in patients in those with poor GCS, while the plasma levels have been seen to decrease after 36 h among survivors [82, 131, 132, 133]. On the other hand, S100B protein has been suggested as a tool for monitoring management efficacy [134, 135], since it has been seen that the blood concentrations of the protein decrease after effective neurosurgical treatment thus making its role more relevant. A high level of S100B during the initial TBI can predict a poor outcome, especially if it is accompanied by a second increase in levels of serum S100B that occurs during the subacute phase [131, 136]. This second peak during the subacute phase may be due to secondary injury to the astroglial cells exhibiting excitotoxicity and neuroinflammation. In addition, elevation in serum levels of S100B and GFAP in TBI patients has been correlated with unfavorable neurological outcomes [137, 138, 139]. On the other hand, an initial lower level of S100B and the lack of second peak might suggest the occurrence of a mild TBI and a good functional recovery [140, 141].

A previous study demonstrated the sensitivity of S100B to predict significant intracranial pathology up to 100% but with specificity of only 28%.Moreover, in pediatric population (specifically for children under the age of 2 years), S100B is not a useful marker due to high normal levels in this group [32, 142]. Thus, S100B may be suggested to be used as an adjuvant marker in patients with TBI, but its diagnostic value is still controversial [76].

Advertisement

5. Limitations of the existing biomarkers

Doctors in the acute hospital settings primarily rely on the patient’s neurological examinations and radiologic imaging to characterize TBI/DAI diagnosis. Depending on the severity of the initial insult, different imaging modalities such as CT scan and MRI are used to obtain the necessary information for patient care and prognosis. However, CT scans, used for assessing cerebrovascular integrity or for determining gross anatomical changes induced to the brain, have low sensitivity to diffuse brain damage, and confers exposure to radiation [143]. In contrast, while MRI can provide information on the extent of diffuse injuries, yet its widespread application is restricted by prohibitive cost, limited availability of MRI in many hospitals, and practical difficulty of performing it in physiologically unstable TBI patients [143]. Thus, diagnostic and prognostic tools for risk stratification of TBI patients are very limited in the early stages after injury.

To fill this gap, research in the field of biofluid-based TBI biomarkers has increased exponentially over the last three decades [116]. Extensive research on fluid biomarkers have demonstrated that a number of brain-specific proteins, as illustrated above, have potential for acting as biomarkers of TBI. These biomolecules are released into the CSF and/or blood, after brain injury due to damage of neural cells [28, 144, 145, 146]. Additionally, neuroimmune activation might have the potential to be novel diagnostic and/or prognostic marker of TBI. A few of these molecules, like S100B have shown promise to be clinically used as biomarkers of TBI [145]. However, this has been disputed in recent studies [147] and till now, there are no rapid, definitive diagnostic blood tests for TBI.

Despite its high sensitivity and negative predictive value, S100B protein is not a specific marker of the CNS damage. Polytraumatized patients without TBI can present S100B protein elevations in blood, though the concentrations return to normal within 6 h after trauma. Patients with brain damage and associated extracranial injuries (hypotension, hypothermia, coagulopathy, inotropic drugs, sedatives, corticosteroids, etc.) can alter the early assessment of S100B protein. Therefore, early determination of this protein is to be avoided in patients with extracranial injuries associated with TBI making it’s role dismal probably in trauma care even though the above features of an ideal biomarker are met.

5.1 Difficulty with interpretation

CNS is very complex and can present a range of different lesions, which in turn can affect different target cells with variable degrees of severity. Brain damage markers must establish differentiations with respect to other alterations. Furthermore, the existence of the blood–brain barrier conditions the structural characteristics of these biomarkers, which must be able to cross the mentioned barrier in order to reach the bloodstream. Biomarkers are dynamic elements that experience changes in response to different inflammatory states, tissue necrosis etc. So serial measurements rather than isolated or point determinations are thus required.

5.2 Controversy

As direct sampling of the damaged brain tissue is not practically feasible there is some controversy regarding the type of biological fluid that should be analyzed. CSF compartment is located closer to the damage site, but frequent collection of CSF samples is unethical. As a result, most biomarkers are studied in peripheral blood as the process is simple, accessible and reproducible. Thus, estimation of blood biomarkers will be the most appropriate option for performing simple and minimally invasive serial measurements. Still more easy will be to estimate biomarkers in fluids that serve as vehicles for their clearance, for example urine.

6. Newly discovered biomarkers of interest.

Advertisement

6.1 Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1)

UCH-L1 mainly resides in the cytoplasm of neuronal cell body representing approximately 5% of all the soluble brain proteins. It is implicated in the elimination of degraded and denatured proteins following oxidative phenomena [148]. Proteomics data first implicate UCH-L1 as promising TBI biomarker candidate [149, 150]. Later studies point to it as a promising brain damage biomarker, since there are data indicating that it can predict the presence of lesions on the CT scan, the need for neurosurgery, and the outcome of patients with TBI [151, 152, 153].

UCH-L1 can be detected in blood, with early increases in its serum concentration following brain injury [151]. The protein level in the blood is shown to be elevated both in mild and severe cases of TBI [100, 154]. Mondello et al. (2012) have obtained interesting results regarding its possible capacity to distinguish between focal and diffuse brain damage [155]. Additionally, it has been suggested that UCH-L1 together with GFAP form the foundation of a biomarker panel representing the two dominant cell types (neuron and astrocytes) in the brain [38]. Interestingly, serum levels of both of these proteins are elevated in professional breacher trainees who were exposed to repeated explosive discharges as well as athletes who experienced concussions [39, 40]. Further investigations are needed to evaluate the properties of this protein as a promising biomarker of DAI.

Advertisement

7. Spectrin degradation products (SBDPs)

Spectrin is a cytoskeletal protein that lines the intracellular side of the plasma membrane forming a scaffold, which maintains plasma membrane integrity and cytoskeletal structure [156]. It is a heterodimeric protein, composed of two α and two β chains, and contains 106 contiguous amino acid sequence motifs called “spectrin repeats”, which are essential to diverse cell functions such as cell adhesion, cell spreading, and the cell cycle [156].

Necrotic and apoptotic cell death during primary and secondary brain injury respectively, cause overactivation of cysteine proteases, such as calpain and caspase-3. These proteases cleave components of the axonal cytoskeleton [157] including spectrin resulting in generation of SBDPs with characteristic molecular weights [26, 158]. The presence of degradation products of spectrin has been described in the CNS in axons and presynaptic neuronal endings [23, 44, 159, 160]. However, SBDPs are not brain specific and its increased serum levels may reflect multiorgan damage in trauma [42, 161]. Moreover, accurate quantification of brain-derived SBDPs in blood is difficult since some proteins found in erythrocytes are similar to those found in the neuronal cytoskeleton [162], thus reducing the diagnostic value of SBDPs.

Advertisement

8. Neurofilament light chain (NfL)

Neurofilaments, consisting of three chains, light (L), medium (M) and heavy (H), make up part of the axonal cytoskeleton. NfL is 68 kDa subunit of the neurofilaments located on the neuronal cytoplasm which is released in response to CNS neuronal damage due to neuroinflammation, neurodegeneration, and/or traumatic or vascular injury [163, 164]. Following axon damage, the influx of calcium alters the phosphorylation state of NfL and subsequent proteolysis. As a result, there is loss of cytoskeletal structure and NfL is released into both CSF and the bloodstream [165]. A number of investigational studies have underscored the role of NfL as biomarker of axon damage [45, 166, 167]. Serum NfL has been shown to distinguish patients with mild, moderate or severe TBI for months and even years after injury [168]. However, serum detection of NfH is considered a better biomarker candidate [169].

Advertisement

9. Conclusions

Since all brain damage biomarkers have some limitation precluding their universal application in the management of severe TBI/DAI, they do not yet form part of routine clinical practice. Some markers, such as NSE and S100B protein, have shown good correlations to clinical severity, the extent of brain damage, response to treatment, and patient outcome. However, the limitations associated with the clinical yield of the molecule or invasiveness of the technique required to obtain the sample have not allowed their generalized use in this patient population.

On the other hand, further studies are needed to understand the role of these proteins in the physiology of the CNS and in the physiopathology of severe TBI/DAI, as well as to clarify the usefulness of those biomarkers that appear to be promising in this field. In this respect, mention must be made of nervous tissue-specific GFAP, as well as of other biomarkers that are currently the focus of interest, such as ubiquitin carboxy-terminal hydrolase L1, the light neurofilaments and spectrin degradation products.

Since these molecules offer isolated information on some of the many elements implicated in the physiopathology of TBI/DAI, we believe that the best strategy is to analyze them in combination. Rather than seeking a biomarker exclusive for brain damage, this approach would allow us to define a panel of biomarkers which jointly – and considering the characteristics inherent to each of them–could offer information referred to severity, the potential benefits of management, and the evolutive course of patients following severe TBI. Only in this way can we hope to complement the traditional methods with a tool that is simple, non-invasive, reproducible and extraordinarily useful for addressing and managing severe TBI/DAI. Moreover, since TBI/DAI is quite complex heterogeneous conditions, it might be clinically justified to use multi-modal biomarkers to evaluate the status of full clinical endophenotypes by combining a panel of biofluid-based and physiologic biomarkers coupled with advanced neuroimaging that are appropriately obtained at multiple time points during the time course of TBI/DAI.

Advertisement

Conflicts of interest

The authors declare that they have no conflicts of interest.

Financial support

RRM is a senior research fellow of Indian Council of Medical Research (ICMR), Government of India. GC is supported by The Ramalingaswami fellowship from the Department of Biotechnology, and grants from the Department of Health Research and ICMR, Government of India.

References

  1. 1. Werner C, Engelhard K. Pathophysiology of traumatic brain injury. British Journal of Anaesthesia. 2007;99:4-9. DOI: 10.1093/BJA/AEM131
  2. 2. Hoffman SW, Harrison C. The interaction between psychological health and traumatic brain injury: A neuroscience perspective. The Clinical Neuropsychologist. 2009;23:1400-1415. DOI: 10.1080/13854040903369433
  3. 3. Kalra S, Banderwal R, Arora K, Kumar S, Singh G, Chawla PA, et al. An update on pathophysiology and treatment of sports-mediated brain injury. Environmental Science and Pollution Research International. 2022;29:16786-16798. DOI: 10.1007/S11356-021-18391-5
  4. 4. Center for Disease Control. Injury Prevention and Control: Traumatic Brain Injury – Concussion. n.d. Available from: https://www.google.com/search?q=CDC%2C+2022.+Injury+Prevention+and+Control%3A+Traumatic+Brain+Injury+Center+for+Disease+Control+(CDC)+and+Prevention.&rlz=1C1YQLS_enIN892IN892&oq=CDC%2C+2022.+Injury+Prevention+and+Control%3A+Traumatic+Brain+Injury+Center+f [Accessed: February 9, 2022]
  5. 5. Bell JM, Breiding MJ, DePadilla L. CDC’s efforts to improve traumatic brain injury surveillance. Journal of Safety Research. 2017;62:253-256. DOI: 10.1016/J.JSR.2017.04.002
  6. 6. Mesfin FB, Gupta N, Shapshak AH, Taylor RS. Diffuse axonal injury. Encycl. Neurological Sciences. 2021:10-12. DOI: 10.1016/b0-12-226870-9/00726-7
  7. 7. Wilson L, Stewart W, Dams-O’Connor K, Diaz-Arrastia R, Horton L, Menon DK, et al. The chronic and evolving neurological consequences of traumatic brain injury. Lancet Neurology. 2017;16:813-825. DOI: 10.1016/S1474-4422(17)30279-X
  8. 8. Huffman JC, Brennan MM, Smith FA, Stern TA. 19 – Patients with neurologic conditions I. seizure disorders (including nonepileptic seizures), cerebrovascular disease, and traumatic brain injury. Massachusetts General Hospital Handbook of General Hospital Psychiatry, Elsevier. 2010:237-253. DOI: 10.1016/B978-1-4377-1927-7.00019-4
  9. 9. Skandsen T, Kvistad KA, Solheim O, Strand IH, Folvik M, Anne V. 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: Clinical article. Journal of Neurosurgery. 2010;113:556-563. DOI: 10.3171/2009.9.JNS09626
  10. 10. Ng SY, Lee AYW. Traumatic brain injuries: Pathophysiology and potential therapeutic targets. Frontiers in Cellular Neuroscience. 2019;13:528. DOI: 10.3389/FNCEL.2019.00528
  11. 11. Schmidt OI, Morganti-Kossmann MC, Heyde CE, Perez D, Yatsiv I, Shohami E, et al. Tumor necrosis factor-mediated inhibition of interleukin-18 in the brain: A clinical and experimental study in head-injured patients and in a murine model of closed head injury. Journal of Neuroinflammation. 2004;1:1-6. DOI: 10.1186/1742-2094-1-13
  12. 12. Ganju A. Intensive care in neurosurgery. The Journal of Trauma and Acute Care Surgery. 2003;54:798. DOI: 10.1097/01.ta.0000058928.27063.6e
  13. 13. Ray SK, Dixon CE, Banik NL. Molecular mechanisms in the pathogenesis of traumatic brain injury. Histology and Histopathology. 2002;17:1137-1152. DOI: 10.14670/HH-17.1137
  14. 14. Smith DH, Meaney DF, Shull WH. Diffuse axonal injury in head trauma. The Journal of Head Trauma Rehabilitation. 2003;18:307-316. DOI: 10.1097/00001199-200307000-00003
  15. 15. Philippi CL, Velez CS, Wade BSC, Drennon AM, Cooper DB, Kennedy JE, et al. Distinct patterns of resting-state connectivity in U.S. service members with mild traumatic brain injury versus posttraumatic stress disorder. Brain Imaging and Behavior. 2021;15:2616-2626. DOI: 10.1007/S11682-021-00464-1
  16. 16. Zhang J, Puvenna V, Janigro D. Biomarkers of traumatic brain injury and their relationship to pathology. In: Translational Research in Traumatic Brain Injury. Boca Raton (FL): CRC Press; 2016. pp. 263-276. DOI: 10.1201/b18959-17
  17. 17. Chatfield DA, Zemlan FP, Day DJ, Menon DK. Discordant temporal patterns of S100beta and cleaved tau protein elevation after head injury: A pilot study. British Journal of Neurosurgery. 2002;16:471-476. DOI: 10.1080/0268869021000030285
  18. 18. Rubenstein R, Chang B, Yue JK, Chiu A, Winkler EA, Puccio AM, et al. Comparing plasma phospho tau, total tau, and phospho tau-total tau ratio as acute and chronic traumatic brain injury biomarkers. JAMA Neurology. 2017;74:1063-1072. DOI: 10.1001/JAMANEUROL.2017.0655
  19. 19. Liliang PC, Liang CL, Lu K, Wang KW, Weng HC, Hsieh CH, et al. Relationship between injury severity and serum tau protein levels in traumatic brain injured rats. Resuscitation. 2010;81:1205-1208. DOI: 10.1016/J.RESUSCITATION.2010.05.016
  20. 20. Du X, Wang X, Geng M. Alzheimer’s disease hypothesis and related therapies. Translational Neurodegeneration. 2018;7:1-7. DOI: 10.1186/s40035-018-0107-y
  21. 21. Franz G, Beer R, Kampfl A, Engelhardt K, Schmutzhard E, Ulmer H, et al. Amyloid beta 1-42 and tau in cerebrospinal fluid after severe traumatic brain injury. Neurology. 2003;60:1457-1461. DOI: 10.1212/01.WNL.0000063313.57292.00
  22. 22. Emmerling MR, Morganti-Kossmann MC, Kossmann T, Stahel PF, Watson MD, Evans LM, et al. Traumatic brain injury elevates the Alzheimer’s amyloid peptide Aβ42 in human CSF. A possible role for nerve cell injury. Annals of the New York Academy of Sciences. 2000;903:118-122. DOI: 10.1111/J.1749-6632.2000.TB06357.X
  23. 23. Ringger NC, O’Steen BE, Brabham JG, Silver X, Pineda J, Wang KKW, et al. A novel marker for traumatic brain injury: CSF alphaII-spectrin breakdown product levels. Journal of Neurotrauma. 2004;21:1443-1456. DOI: 10.1089/NEU.2004.21.1443
  24. 24. Zhang Z, Moghieb A, Wang KKW. Neuro-proteomics and neuro-systems biology in the quest of TBI biomarker discovery. In: Biomarkers of the Brain Injury and Neurological Disorders. Boca Raton (FL): CRC Press; 2014. pp. 21-59. DOI: 10.1201/b17644-7
  25. 25. Thompson RJ, Kynoch PAM, Sarjant J. Immunohistochemical localization of creatine kinase-BB isoenzyme to astrocytes in human brain. Brain Research. 1980;201:423-426. DOI: 10.1016/0006-8993(80)91046-X
  26. 26. Toman E, Harrisson S, Belli T. Biomarkers in traumatic brain injury: A review. Journal of the Royal Army Medical Corps. 2016;162:103-108. DOI: 10.1136/JRAMC-2015-000517
  27. 27. Ingebrigtsen T, Romner B. Biochemical serum markers of traumatic brain injury. The Journal of Trauma. 2002;52:798-808. DOI: 10.1097/00005373-200204000-00038
  28. 28. Buonora JE, Yarnell AM, Lazarus RC, Mousseau M, Latour LL, Rizoli SB, et al. Multivariate analysis of traumatic brain injury: Development of an assessment score. Frontiers in Neurology. 2015;6:68. DOI: 10.3389/FNEUR.2015.00068
  29. 29. Böhmer AE, Oses JP, Schmidt AP, Perón CS, Krebs CL, Oppitz PP, et al. Neuron-specific enolase, S100B, and glial fibrillary acidic protein levels as outcome predictors in patients with severe traumatic brain injury. Neurosurgery. 2011;68:1624-1630. DOI: 10.1227/NEU.0B013E318214A81F
  30. 30. Stein DM, Kufera JA, Lindell A, Murdock KR, Menaker J, Bochicchio GV, et al. Association of CSF biomarkers and secondary insults following severe traumatic brain injury. Neurocritical Care. 2011;14:200-207. DOI: 10.1007/S12028-010-9496-1
  31. 31. Topolovec-Vranic J, Pollmann-Mudryj MA, Ouchterlony D, Klein D, Spence J, Romaschin A, et al. The value of serum biomarkers in prediction models of outcome after mild traumatic brain injury. The Journal of Trauma. 2011;71:S478-S486. DOI: 10.1097/TA.0B013E318232FA70
  32. 32. Berger RP, Dulani T, Adelson PD, Leventhal JM, Richichi R, Kochanek PM. Identification of inflicted traumatic brain injury in well-appearing infants using serum and cerebrospinal markers: A possible screening tool. Pediatrics. 2006;117:325-332. DOI: 10.1542/PEDS.2005-0711
  33. 33. Gonçalves CA, Concli Leite M, Nardin P. Biological and methodological features of the measurement of S100B, a putative marker of brain injury. Clinical Biochemistry. 2008;41:755-763. DOI: 10.1016/J.CLINBIOCHEM.2008.04.003
  34. 34. Calcagnile O, Undén L, Undén J. Clinical validation of S100B use in management of mild head injury. BMC Emergency Medicine. 2012;12:1-6. DOI: 10.1186/1471-227X-12-13
  35. 35. Czeiter E, Mondello S, Kovacs N, Sandor J, Gabrielli A, Schmid K, et al. Brain injury biomarkers may improve the predictive power of the IMPACT outcome calculator. Journal of Neurotrauma. 2012;29:1770. DOI: 10.1089/NEU.2011.2127
  36. 36. Mondello S, Kobeissy F, Vestri A, Hayes RL, Kochanek PM, Berger RP. Serum concentrations of ubiquitin C-terminal hydrolase-L1 and glial fibrillary acidic protein after pediatric traumatic brain injury. Scientific Reports. 2016;6:1-8. DOI: 10.1038/SREP28203
  37. 37. Davidoff MS, Middendorff R, Köfüncü E, Müller D, Ježek D, Holstein AF. Leydig cells of the human testis possess astrocyte and oligodendrocyte marker molecules. Acta Histochemica. 2002;104:39-49. DOI: 10.1078/0065-1281-00630
  38. 38. Welch RD, Ayaz SI, Lewis LM, Unden J, Chen JY, Mika VH, et al. Ability of serum glial fibrillary acidic protein, ubiquitin C-terminal hydrolase-L1, and S100B to differentiate normal and abnormal head computed tomography findings in patients with suspected mild or moderate traumatic brain injury. Journal of Neurotrauma. 2016;33:203. DOI: 10.1089/NEU.2015.4149
  39. 39. Tate CM, Wang KKW, Eonta S, Zhang Y, Carr W, Tortella FC, et al. Serum brain biomarker level, neurocognitive performance, and self-reported symptom changes in soldiers repeatedly exposed to low-level blast: A breacher pilot study. Journal of Neurotrauma. 2013;30:1620-1630. DOI: 10.1089/NEU.2012.2683
  40. 40. Puvenna V, Brennan C, Shaw G, Yang C, Marchi N, Bazarian JJ, et al. Significance of ubiquitin carboxy-terminal hydrolase L1 elevations in athletes after sub-concussive head hits. PLoS One. 2014;9:e96296. DOI: 10.1371/JOURNAL.PONE.0096296
  41. 41. Zoltewicz JS, Mondello S, Yang B, Newsom KJ, Kobeissy F, Yao C, et al. Biomarkers track damage after graded injury severity in a rat model of penetrating brain injury. Journal of Neurotrauma. 2013;30:1161-1169. DOI: 10.1089/NEU.2012.2762
  42. 42. Yan X-X, Jeromin A. Spectrin breakdown products (SBDPs) as potential biomarkers for neurodegenerative diseases. Current Translational Geriatrics and Experimental Gerontology Reports. 2012;1:85-93. DOI: 10.1007/s13670-012-0009-2
  43. 43. Mondello S, Robicsek SA, Gabrielli A, Brophy GM, Papa L, Tepas J, et al. αII-Spectrin breakdown products (SBDPs): Diagnosis and outcome in severe traumatic brain injury patients. Journal of Neurotrauma. 2010;27:1203. DOI: 10.1089/NEU.2010.1278
  44. 44. Pineda JA, Lewis SB, Valadka AB, Papa L, Hannay HJ, Heaton SC, et al. Clinical significance of alphaII-spectrin breakdown products in cerebrospinal fluid after severe traumatic brain injury. Journal of Neurotrauma. 2007;24:354-366. DOI: 10.1089/NEU.2006.003789
  45. 45. Sass D, Guedes VA, Smith EG, Vorn R, Devoto C, Edwards KA, et al. Sex differences in behavioral symptoms and the levels of circulating GFAP, tau, and NfL in patients with traumatic brain injury. Frontiers in Pharmacology. 2021;12:3367. DOI: 10.3389/FPHAR.2021.746491
  46. 46. Khatoon S, Grundke-Iqbal I, Iqbal K. Levels of normal and abnormally phosphorylated tau in different cellular and regional compartments of Alzheimer disease and control brains. FEBS Letters. 1994;351:80-84. DOI: 10.1016/0014-5793(94)00829-9
  47. 47. Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harbor Perspectives in Medicine. 2012;2:1-25. DOI: 10.1101/CSHPERSPECT.A006247
  48. 48. Duan Y, Dong S, Gu F, Hu Y, Zhao Z. Advances in the pathogenesis of Alzheimer’s disease: Focusing on tau-mediated neurodegeneration. Translational Neurodegeneration. 2012;1:24. DOI: 10.1186/2047-9158-1-24
  49. 49. Yang Z, Wang P, Morgan D, Lin D, Pan J, Lin F, et al. Temporal MRI characterization, neurobiochemical and neurobehavioral changes in a mouse repetitive concussive head injury model. Scientific Reports 2015;5(1):1-15. doi:10.1038/srep11178
  50. 50. Rubenstein R, Chang B, Davies P, Wagner AK, Robertson CS, Wang KKW. A novel, ultrasensitive assay for tau: Potential for assessing traumatic brain injury in tissues and biofluids. Journal of Neurotrauma. 2015;32:342. DOI: 10.1089/NEU.2014.3548
  51. 51. Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harbor Perspectives in Biology. 2011;3:1-25. DOI: 10.1101/cshperspect.a006247
  52. 52. Iqbal K, Grundke-Iqbal I. Ubiquitination and abnormal phosphorylation of paired helical filaments in Alzheimer’s disease. Molecular Neurobiology. 1991;5:399-410. DOI: 10.1007/BF02935561
  53. 53. Moretti L, Cristofori I, Weaver SM, Chau A, Portelli JN, Grafman J. Cognitive decline in older adults with a history of traumatic brain injury. Lancet Neurology. 2012;11:1103-1112. DOI: 10.1016/S1474-4422(12)70226-0
  54. 54. Sivanandam TM, Thakur MK. Traumatic brain injury: A risk factor for Alzheimer’s disease. Neuroscience and Biobehavioral Reviews. 2012;36:1376-1381. DOI: 10.1016/J.NEUBIOREV.2012.02.013
  55. 55. Lee PC, Bordelon Y, Bronstein J, Ritz B. Traumatic brain injury, paraquat exposure, and their relationship to Parkinson disease. Neurology. 2012;79:2061-2066. DOI: 10.1212/WNL.0B013E3182749F28
  56. 56. Dams-O’Connor K, Gibbons LE, Bowen JD, McCurry SM, Larson EB, Crane PK. Risk for late-life re-injury, dementia and death among individuals with traumatic brain injury: A population-based study. Journal of Neurology, Neurosurgery, and Psychiatry. 2013;84:177-182. DOI: 10.1136/JNNP-2012-303938
  57. 57. Hu W, Tung YC, Zhang Y, Liu F, Iqbal K. Involvement of activation of asparaginyl endopeptidase in tau hyperphosphorylation in repetitive mild traumatic brain injury. Journal of Alzheimer’s Disease. 2018;64:709. DOI: 10.3233/JAD-180177
  58. 58. Marklund N, Vedung F, Lubberink M, Tegner Y, Johansson J, Blennow K, et al. Tau aggregation and increased neuroinflammation in athletes after sports-related concussions and in traumatic brain injury patients - a PET/MR study. NeuroImage: Clinical. 2021;30:102665. DOI: 10.1016/J.NICL.2021.102665
  59. 59. Zemlan FP, Rosenberg WS, Luebbe PA, Campbell TA, Dean GE, Weiner NE, et al. Quantification of axonal damage in traumatic brain injury: Affinity purification and characterization of cerebrospinal fluid tau proteins. Journal of Neurochemistry. 1999;72:741-750. DOI: 10.1046/J.1471-4159.1999.0720741.X
  60. 60. Zemlan FP, Jauch EC, Mulchahey JJ, Gabbita SP, Rosenberg WS, Speciale SG, et al. C-tau biomarker of neuronal damage in severe brain injured patients: Association with elevated intracranial pressure and clinical outcome. Brain Research. 2002;947:131-139. DOI: 10.1016/S0006-8993(02)02920-7
  61. 61. Smith DH, Chen XH, Nonaka M, Trojanowski JQ, Lee VMY, Saatman KE, et al. Accumulation of amyloid beta and tau and the formation of neurofilament inclusions following diffuse brain injury in the pig. Journal of Neuropathology and Experimental Neurology. 1999;58:982-992. DOI: 10.1097/00005072-199909000-00008
  62. 62. Pandey S, Singh K, Sharma V, Pandey D, Jha RP, Rai SK, et al. A prospective pilot study on serum cleaved tau protein as a neurological marker in severe traumatic brain injury. British Journal of Neurosurgery. 2017;31:356-363. DOI: 10.1080/02688697.2017.1297378
  63. 63. Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet (London, England). 2011;377:1019-1031. DOI: 10.1016/S0140-6736(10)61349-9
  64. 64. Wang J, Gu BJ, Masters CL, Wang YJ. A systemic view of Alzheimer disease - insights from amyloid-β metabolism beyond the brain. Nature Reviews. Neurology. 2017;13:612-623. DOI: 10.1038/nrneurol.2017.111
  65. 65. Liu PP, Xie Y, Meng XY, Kang JS. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduction and Targeted Therapy. 2019;4(1):1-22. DOI: 10.1038/s41392-019-0063-8
  66. 66. Scheltens P, Blennow K, Breteler MMB, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer’s disease. Lancet (London, England). 2016;388:505-517. DOI: 10.1016/S0140-6736(15)01124-1
  67. 67. Xu J, Rasmussen IA, Lagopoulos J, Haberg A. Diffuse axonal injury in severe traumatic brain injury visualized using high-resolution diffusion tensor imaging. Journal of Neurotrauma. 2007;24:753-765. DOI: 10.1089/NEU.2006.0208
  68. 68. Deber CM, Reynolds SJ. Central nervous system myelin: Structure, function, and pathology. Clinical Biochemistry. 1991;24:113-134. DOI: 10.1016/0009-9120(91)90421-A
  69. 69. Ming CL, Akle V, Zheng W, Kitlen J, O’Steen B, Larner SF, et al. Extensive degradation of myelin basic protein isoforms by calpain following traumatic brain injury. Journal of Neurochemistry. 2006;98:700-712. DOI: 10.1111/J.1471-4159.2006.03882.X
  70. 70. Ottens AK, Golden EC, Bustamante L, Hayes RL, Denslow ND, Wang KKW. Proteolysis of multiple myelin basic protein isoforms after neurotrauma: Characterization by mass spectrometry. Journal of Neurochemistry. 2008;104:1404-1414. DOI: 10.1111/J.1471-4159.2007.05086.X
  71. 71. Thomas DGT, Palfreyman JW, Ratcliffe JG. Serum-myelin-basic-protein assay in diagnosis and prognosis of patients with head injury. Lancet (London, England). 1978;1:113-115. DOI: 10.1016/S0140-6736(78)90415-4
  72. 72. Berger RP, Adelson PD, Pierce MC, Dulani T, Cassidy LD, Kochanek PM. Serum neuron-specific enolase, S100B, and myelin basic protein concentrations after inflicted and noninflicted traumatic brain injury in children. Journal of Neurosurgery. 2005;103:61-68. DOI: 10.3171/PED.2005.103.1.0061
  73. 73. Beers SR, Berger RP, Adelson PD. Neurocognitive outcome and serum biomarkers in inflicted versus non-inflicted traumatic brain injury in young children. Journal of Neurotrauma. 2007;24:97-105. DOI: 10.1089/NEU.2006.0055
  74. 74. Giacoppo S, Bramanti P, Barresi M, Celi D, Cuzzola VF, Palella E, et al. Predictive biomarkers of recovery in traumatic brain injury. Neurocritical Care. 2012;16:470-477. DOI: 10.1007/S12028-012-9707-Z
  75. 75. Yamazaki Y, Ohtaka H, Morii S, Kitahara T, Ohwada T, Yada K. Diagnostic significance of serum neuron-specific enolase and myelin basic protein assay in patients with acute head injury. Surgical Neurology. 1995;43:381-383. DOI: 10.1007/978-4-431-68231-8_86
  76. 76. Mehta T, Fayyaz M, Giler GE, Kaur H, Raikwar SP, Kempuraj D, et al. Current trends in biomarkers for traumatic brain injury. Open Access Journal of Neurology & Neurosurgery. 2020;12:86
  77. 77. Wallimann T, Hemmer W. Creatine kinase in non-muscle tissues and cells. Molecular and Cellular Biochemistry. 1994;133-134:193-220. DOI: 10.1007/BF01267955
  78. 78. Rabow L, Hedman G. CKBB-isoenzymes as a sign of cerebral injury. Acta Neurochirurgica. Supplementum (Wien). 1979;28:108-112. DOI: 10.1007/978-3-7091-4088-8_24
  79. 79. Kaste M, Hernesniemi J, Somer H, Hillbom M, Konttinen A. Creatine kinase isoenzymes in acute brain injury. Journal of Neurosurgery. 1981;55:511-515. DOI: 10.3171/JNS.1981.55.4.0511
  80. 80. Rabow L, DeSalles AAF, Becker DP, Yang M, Kontos HA, Ward JD, et al. CSF brain creatine kinase levels and lactic acidosis in severe head injury. Journal of Neurosurgery. 1986;65:625-629. DOI: 10.3171/JNS.1986.65.5.0625
  81. 81. Skogseid IM, Nordby HK, Urdal P, Paus E, Lilleaas F. Increased serum creatine kinase BB and neuron specific enolase following head injury indicates brain damage. Acta Neurochirurgica. 1992;115:106-111. DOI: 10.1007/BF01406367
  82. 82. Mussack T, Kirchhoff C, Buhmann S, Biberthaler P, Ladurner R, Gippner-Steppert C, et al. Significance of Elecsys S100 immunoassay for real-time assessment of traumatic brain damage in multiple trauma patients. Clinical Chemistry and Laboratory Medicine. 2006;44:1140-1145. DOI: 10.1515/CCLM.2006.190
  83. 83. Stopfkuchen H, Schranz D, Salzmann G, Jüngst BK, Wiss M, Schwarz M. The diagnostic value of CK-MB isoenzyme determinations in children with head injury and polytrauma. Monatsschrift für Kinderheilkunde. 1984;132:594-599
  84. 84. Kärkelä J, Bock E, Kaukinen S. CSF and serum brain-specific creatine kinase isoenzyme (CK-BB), neuron-specific enolase (NSE) and neural cell adhesion molecule (NCAM) as prognostic markers for hypoxic brain injury after cardiac arrest in man. Journal of the Neurological Sciences. 1993;116:100-109. DOI: 10.1016/0022-510X(93)90095-G
  85. 85. Morris MC, Bercz A, Niziolek GM, Kassam F, Veile R, Friend LA, et al. UCH-L1 is a poor serum biomarker of murine traumatic brain injury after polytrauma. The Journal of Surgical Research. 2019;244:63-68. DOI: 10.1016/J.JSS.2019.06.023
  86. 86. Berger RP, Pierce MC, Wisniewski SR, Adelson PD, Clark RSB, Ruppel RA, et al. Neuron-specific enolase and S100B in cerebrospinal fluid after severe traumatic brain injury in infants and children. Pediatrics. 2002;109:E31. DOI: 10.1542/PEDS.109.2.E31
  87. 87. Liu M, Zhang C, Liu W, Luo P, Zhang L, Wang Y, et al. A novel rat model of blast-induced traumatic brain injury simulating different damage degree: Implications for morphological, neurological, and biomarker changes. Frontiers in Cellular Neuroscience. 2015;9:168. DOI: 10.3389/FNCEL.2015.00168
  88. 88. Verfaillie CJ, Delanghe JR. Hemolysis correction factor in the measurement of serum neuron-specific enolase. Clinical Chemistry and Laboratory Medicine. 2010;48:891-892. DOI: 10.1515/CCLM.2010.159
  89. 89. Pfeifer R, Börner A, Krack A, Sigusch HH, Surber R, Figulla HR. Outcome after cardiac arrest: Predictive values and limitations of the neuroproteins neuron-specific enolase and protein S-100 and the Glasgow coma scale. Resuscitation. 2005;65:49-55. DOI: 10.1016/J.RESUSCITATION.2004.10.011
  90. 90. Pelinka LE, Jafarmadar M, Redl H, Bahrami S. Neuron-specific-enolase is increased in plasma after hemorrhagic shock and after bilateral femur fracture without traumatic brain injury in the rat. Shock. 2004;22:88-91. DOI: 10.1097/01.SHK.0000130157.34382.3F
  91. 91. Wolf H, Krall C, Pajenda G, Leitgeb J, Bukaty AJ, Hajdu S, et al. Alterations of the biomarker S-100B and NSE in patients with acute vertebral spine fractures. The Spine Journal. 2014;14:2918-2922. DOI: 10.1016/j.spinee.2014.04.027
  92. 92. Kok WF, Koerts J, Tucha O, Scheeren TWL, Absalom AR. Neuronal damage biomarkers in the identification of patients at risk of long-term postoperative cognitive dysfunction after cardiac surgery. Anaesthesia. 2017;72:359-369. DOI: 10.1111/ANAE.13712
  93. 93. Yuan X, Wang J, Wang D, Yang S, Yu N, Guo F. NSE, S100B and MMP9 expression following reperfusion after carotid artery stenting. Current Neurovascular Research. 2019;16:129-134. DOI: 10.2174/1567202616666190321123515
  94. 94. Zhang X, Ma S, Chen Y, Yin Y, Bai W, Tan J, et al. The isocitrate dehydrogenase 1 is a potential prognostic indicator for non-small cell lung cancer patients. The International Journal of Biological Markers. 2021;36:27-35. DOI: 10.1177/17246008211052571
  95. 95. Missler U, Orlowski N, Nötzold A, Dibbelt L, Steinmeier E, Wiesmann M. Early elevation of S-100B protein in blood after cardiac surgery is not a predictor of ischemic cerebral injury. Clinica Chimica Acta. 2002;321:29-33. DOI: 10.1016/S0009-8981(02)00061-X
  96. 96. Okonkwo DO, Yue JK, Puccio AM, Panczykowski DM, Inoue T, McMahon PJ, et al. GFAP-BDP as an acute diagnostic marker in traumatic brain injury: Results from the prospective transforming research and clinical knowledge in traumatic brain injury study. Journal of Neurotrauma. 2013;30:1490-1497. DOI: 10.1089/NEU.2013.2883
  97. 97. Yang Z, Wang KKW. Glial fibrillary acidic protein: From intermediate filament assembly and gliosis to neurobiomarker. Trends in Neurosciences. 2015;38:364-374. DOI: 10.1016/J.TINS.2015.04.003
  98. 98. Adrian H, Mårten K, Salla N, Lasse V. Biomarkers of traumatic brain injury: Temporal changes in body fluids. ENeuro. 2016;3:ENEURO.0294-16.2016. DOI: 10.1523/ENEURO.0294-16.2016
  99. 99. Nylén K, Öst M, Csajbok LZ, Nilsson I, Blennow K, Nellgård B, et al. Increased serum-GFAP in patients with severe traumatic brain injury is related to outcome. Journal of the Neurological Sciences. 2006;240:85-91. DOI: 10.1016/J.JNS.2005.09.007
  100. 100. Diaz-Arrastia R, Wang KKW, Papa L, Sorani MD, Yue JK, Puccio AM, et al. Acute biomarkers of traumatic brain injury: Relationship between plasma levels of ubiquitin C-terminal hydrolase-L1 and glial fibrillary acidic protein. Journal of Neurotrauma. 2014;31:19-25. DOI: 10.1089/NEU.2013.3040
  101. 101. Hainfellner JA, Voigtländer T, Ströbel T, Mazal PR, Maddalena AS, Aguzzi A, et al. Fibroblasts can express glial fibrillary acidic protein (GFAP) in vivo. Journal of Neuropathology and Experimental Neurology. 2001;60:449-461. DOI: 10.1093/JNEN/60.5.449
  102. 102. Posti JP, Hossain I, Takala RSK, Liedes H, Newcombe V, Outtrim J, et al. Glial fibrillary acidic protein and ubiquitin C-terminal hydrolase-L1 are not specific biomarkers for mild CT-negative traumatic brain injury. Journal of Neurotrauma. 2017;34:1427-1438. DOI: 10.1089/NEU.2016.4442
  103. 103. Shemilt M, Boutin A, Lauzier F, Zarychanski R, Moore L, McIntyre LA, et al. Prognostic value of glial fibrillary acidic protein in patients with moderate and severe traumatic brain injury: A systematic review and meta-analysis. Critical Care Medicine. 2019;47:e522-e529. DOI: 10.1097/CCM.0000000000003728
  104. 104. Lei J, Gao G, Feng J, Jin Y, Wang C, Mao Q, et al. Glial fibrillary acidic protein as a biomarker in severe traumatic brain injury patients: A prospective cohort study. Critical Care. 2015;19:1-12. DOI: 10.1186/S13054-015-1081-8/TABLES/3
  105. 105. Fraser DD, Close TE, Rose KL, Ward R, Mehl M, Farrell C, et al. Severe traumatic brain injury in children elevates glial fibrillary acidic protein in cerebrospinal fluid and serum. Pediatric Critical Care Medicine. 2011;12:319-324. DOI: 10.1097/PCC.0B013E3181E8B32D
  106. 106. Olsson B, Zetterberg H, Hampel H, Blennow K. Biomarker-based dissection of neurodegenerative diseases. Progress in Neurobiology. 2011;95:520-534. DOI: 10.1016/J.PNEUROBIO.2011.04.006
  107. 107. Persson L, Hårdemark HG, Gustafsson J, Rundström G, Mendel-Hartvig I, Esscher T, et al. S-100 protein and neuron-specific enolase in cerebrospinal fluid and serum: Markers of cell damage in human central nervous system. Stroke. 1987;18:911-918. DOI: 10.1161/01.STR.18.5.911
  108. 108. Xiong H, Liang WL, Wu XR. Pathophysiological alterations in cultured astrocytes exposed to hypoxia/reoxygenation. Sheng Li Ke Xue Jin Zhan. 2000;31:217-221
  109. 109. Donato R, Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, et al. S100B’s double life: Intracellular regulator and extracellular signal. Biochimica et Biophysica Acta. 2009;1793:1008-1022. DOI: 10.1016/J.BBAMCR.2008.11.009
  110. 110. Michetti F, D’Ambrosi N, Toesca A, Puglisi MA, Serrano A, Marchese E, et al. The S100B story: From biomarker to active factor in neural injury. Journal of Neurochemistry. 2019;148:168-187. DOI: 10.1111/JNC.14574
  111. 111. Einav S, Shoshan Y, Ovadia H, Matot I, Hersch M, Itshayek E. Early postoperative serum S100β levels predict ongoing brain damage after meningioma surgery: A prospective observational study. Critical Care. 2006;10:1-10. DOI: 10.1186/CC5058/FIGURES/4
  112. 112. Anczykowski G, Kaczmarek J, Jankowski R, Guzniczak P. The reference level of serum S-100B protein for poor prognosis in patients with intracranial extracerebral hematoma. EJIFCC. 2011;22:66
  113. 113. Grevfors N, Lindblad C, Nelson DW, Svensson M, Thelin EP, Rubenson WR. Delayed neurosurgical intervention in traumatic brain injury patients referred from primary hospitals is not associated with an unfavorable outcome. Frontiers in Neurology. 2021;11:1799. DOI: 10.3389/FNEUR.2020.610192
  114. 114. Chen YH, Cheng ZY, Shao LH, Shentu HS, Fu B. Macrophage migration inhibitory factor as a serum prognostic marker in patients with aneurysmal subarachnoid hemorrhage. Clinica Chimica Acta. 2017;473:60-64. DOI: 10.1016/J.CCA.2017.08.018
  115. 115. Jönsson H, Johnsson P, Höglund P, Alling C, Blomquist S. Elimination of S100B and renal function after cardiac surgery. Journal of Cardiothoracic and Vascular Anesthesia. 2000;14:698-701. DOI: 10.1053/JCAN.2000.18444
  116. 116. Papa L, Silvestri S, Brophy GM, Giordano P, Falk JL, Braga CF, 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. Journal of Neurotrauma. 2014;31:1815-1822. DOI: 10.1089/NEU.2013.3245
  117. 117. Metting Z, Wilczak N, Rodiger LA, Schaaf JM, Van Der Naalt J. GFAP and S100B in the acute phase of mild traumatic brain injury. Neurology. 2012;78:1428-1433. DOI: 10.1212/WNL.0B013E318253D5C7
  118. 118. Barbosa RR, Jawa R, Watters JM, Knight JC, Kerwin AJ, Winston ES, et al. Evaluation and management of mild traumatic brain injury: An eastern Association for the Surgery of trauma practice management guideline. Journal of Trauma and Acute Care Surgery. 2012;73:S307-S314. DOI: 10.1097/TA.0B013E3182701885
  119. 119. Zongo D, Ribéreau-Gayon R, Masson F, Laborey M, Contrand B, Salmi LR, et al. S100-B protein as a screening tool for the early assessment of minor head injury. Annals of Emergency Medicine. 2012;59:209-218. DOI: 10.1016/J.ANNEMERGMED.2011.07.027
  120. 120. Rothoerl RD, Woertgen C, Holzschuh M, Metz C, Brawanski A. S-100 serum levels after minor and major head injury. The Journal of Trauma. 1998;45:765-767. DOI: 10.1097/00005373-199810000-00025
  121. 121. Herrmann M, Curio N, Jost S, Wunderlich MT, Synowitz H, Wallesch CW. Protein S-100B and neuron specific enolase as early neurobiochemical markers of the severity of traumatic brain injury. Restorative Neurology and Neuroscience. 1999;14:109-114
  122. 122. Biberthaler P, Musaelyan K, Krieg S, Meyer B, Stimmer H, Zapf J, et al. Evaluation of acute glial fibrillary acidic protein and ubiquitin C-terminal hydrolase-L1 plasma levels in traumatic brain injury patients with and without intracranial lesions. Neurotrauma Reports. 2021;2:617. DOI: 10.1089/NEUR.2021.0048
  123. 123. Da Rocha AB, Schneider RF, De Freitas GR, André C, Grivicich I, Zanoni C, et al. Role of serum S100B as a predictive marker of fatal outcome following isolated severe head injury or multitrauma in males. Clinical Chemistry and Laboratory Medicine. 2006;44:1234-1242. DOI: 10.1515/CCLM.2006.218
  124. 124. Rodríguez-Rodríguez A, Egea-Guerrero JJ, León-Justel A, Gordillo-Escobar E, Revuelto-Rey J, Vilches-Arenas Á, et al. Role of S100B protein in urine and serum as an early predictor of mortality after severe traumatic brain injury in adults. Clinica Chimica Acta. 2012;414:228-233. DOI: 10.1016/J.CCA.2012.09.025
  125. 125. Gradisek P, Osredkar J, Korsic M, Kremzar B. Multiple indicators model of long-term mortality in traumatic brain injury. Brain Injury. 2012;26:1472-1481. DOI: 10.3109/02699052.2012.694567
  126. 126. Raheja A, Sinha S, Samson N, Bhoi S, Subramanian A, Sharma P, et al. Serum biomarkers as predictors of long-term outcome in severe traumatic brain injury: Analysis from a randomized placebo-controlled phase II clinical trial. Journal of Neurosurgery. 2016;125:631-641. DOI: 10.3171/2015.6.JNS15674
  127. 127. Thelin EP, Frostell A, Mulder J, Mitsios N, Damberg P, Aski SN, et al. Lesion size is exacerbated in hypoxic rats whereas hypoxia-inducible factor-1 alpha and vascular endothelial growth factor increase in injured normoxic rats: A prospective cohort study of secondary hypoxia in focal traumatic brain injury. Frontiers in Neurology. 2016;7:23. DOI: 10.3389/FNEUR.2016.00023
  128. 128. Rainey T, Lesko M, Sacho R, Lecky F, Childs C. Predicting outcome after severe traumatic brain injury using the serum S100B biomarker: Results using a single (24h) time-point. Resuscitation. 2009;80:341-345. DOI: 10.1016/J.RESUSCITATION.2008.11.021
  129. 129. Egea-Guerrero JJ, Murillo-Cabezas F, Gordillo-Escobar E, Rodríguez-Rodríguez A, Enamorado-Enamorado J, Revuelto-Rey J, et al. S100B protein may detect brain death development after severe traumatic brain injury. Journal of Neurotrauma. 2013;30:1762. DOI: 10.1089/NEU.2012.2606
  130. 130. Kellermann I, Kleindienst A, Hore N, Buchfelder M, Brandner S. Early CSF and serum S100B concentrations for outcome prediction in traumatic brain injury and subarachnoid hemorrhage. Clinical Neurology and Neurosurgery. 2016;145:79-83. DOI: 10.1016/J.CLINEURO.2016.04.005
  131. 131. Thelin EP, Johannesson L, Nelson D, Bellander BM. S100B is an important outcome predictor in traumatic brain injury. Journal of Neurotrauma. 2013;30:519-528. DOI: 10.1089/NEU.2012.2553
  132. 132. Park SH, Hwang SK. Prognostic value of serum levels of S100 calcium-binding protein B, neuron-specific enolase, and interleukin-6 in pediatric patients with traumatic brain injury. World Neurosurgery. 2018;118:e534-e542. DOI: 10.1016/J.WNEU.2018.06.234
  133. 133. Yin W, Weng S, Lai S, Nie H. GCS score combined with CT score and serum S100B protein level can evaluate severity and early prognosis of acute traumatic brain injury. Nan Fang Yi Ke Da Xue Xue Bao. 2021;41:543-548. DOI: 10.12122/J.ISSN.1673-4254.2021.04.09
  134. 134. Korfias S, Stranjalis G, Boviatsis E, Psachoulia C, Jullien G, Gregson B, et al. Serum S-100B protein monitoring in patients with severe traumatic brain injury. Intensive Care Medicine. 2007;33:255-260. DOI: 10.1007/S00134-006-0463-4
  135. 135. Taheri A, Emami M, Asadipour E, Kasirzadeh S, Rouini MR, Najafi A, et al. A randomized controlled trial on the efficacy, safety, and pharmacokinetics of metformin in severe traumatic brain injury. Journal of Neurology. 2019;266:1988-1997. DOI: 10.1007/S00415-019-09366-1
  136. 136. Thelin EP, Nelson DW, Bellander BM. Secondary peaks of S100B in serum relate to subsequent radiological pathology in traumatic brain injury. Neurocritical Care. 2014;20:217-229. DOI: 10.1007/S12028-013-9916-0
  137. 137. Di Battista AP, Buonora JE, Rhind SG, Hutchison MG, Baker AJ, Rizoli SB, et al. Blood biomarkers in moderate-to-severe traumatic brain injury: Potential utility of a multi-marker approach in characterizing outcome. Frontiers in Neurology. 2015;6:110. DOI: 10.3389/FNEUR.2015.00110
  138. 138. Lee JY, Lee CY, Kim HR, Lee CH, Kim HW, Kim JH. A role of serum-based neuronal and glial markers as potential predictors for distinguishing severity and related outcomes in traumatic brain injury. Journal of Korean Neurosurgical Association. 2015;58:93-100. DOI: 10.3340/JKNS.2015.58.2.93
  139. 139. Thelin EP, Nelson DW, Bellander BM. A review of the clinical utility of serum S100B protein levels in the assessment of traumatic brain injury. Acta Neurochirurgica. 2017;159:209-225. DOI: 10.1007/S00701-016-3046-3
  140. 140. Bloomfield SM, McKinney J, Smith L, Brisman J. Reliability of S100B in predicting severity of central nervous system injury. Neurocritical Care. 2007;6:121-138. DOI: 10.1007/S12028-007-0008-X
  141. 141. Pham N, Fazio V, Cucullo L, Teng Q, Biberthaler P, Bazarian JJ, et al. Extracranial sources of S100B do not affect serum levels. PLoS One. 2010;5:e12691. DOI: 10.1371/JOURNAL.PONE.0012691
  142. 142. Piazza O, Storti MP, Cotena S, Stoppa F, Perrotta D, Esposito G, et al. S100B is not a reliable prognostic index in paediatric TBI. Pediatric Neurosurgery. 2007;43:258-264. DOI: 10.1159/000103304
  143. 143. Papa L, Edwards D, Ramia M. Exploring serum biomarkers for mild traumatic brain injury. In: Brain Neurotrauma: Molecular, Neuropsychological, and Rehabilitation Aspects. Boca Raton (FL): CRC Press; 2015. pp. 301-308. DOI: 10.1201/b18126
  144. 144. Blennow K, Zetterberg H. The past and the future of Alzheimer’s disease fluid biomarkers. Journal of Alzheimer’s Disease. 2018;62:1125-1140. DOI: 10.3233/JAD-170773
  145. 145. Li J, Yu C, Sun Y, Li Y. Serum ubiquitin C-terminal hydrolase L1 as a biomarker for traumatic brain injury: A systematic review and meta-analysis. The American Journal of Emergency Medicine. 2015;33:1191-1196. DOI: 10.1016/J.AJEM.2015.05.023
  146. 146. Janigro D, Bailey DM, Lehmann S, Badaut J, O’Flynn R, Hirtz C, et al. Peripheral blood and salivary biomarkers of blood-brain barrier permeability and neuronal damage: Clinical and applied concepts. Frontiers in Neurology. 2021;11:577312. DOI: 10.3389/FNEUR.2020.577312
  147. 147. Rogatzki MJ, Morgan JE, Baker JS, Knox A, Serrador JM. Protein S100B and brain lipid-binding protein concentrations in the serum of recently concussed rugby players. Journal of Neurotrauma. 2021;38:2247-2254. DOI: 10.1089/NEU.2021.0004
  148. 148. Larsen CN, Price JS, Wilkinson KD. Substrate binding and catalysis by ubiquitin C-terminal hydrolases: Identification of two active site residues. Biochemistry. 1996;35:6735-6744. DOI: 10.1021/BI960099F
  149. 149. Kobeissy FH, Ottens AK, Zhang Z, Liu MC, Denslow ND, Dave JR, et al. Novel differential neuroproteomics analysis of traumatic brain injury in rats. Molecular & Cellular Proteomics. 2006;5:1887-1898. DOI: 10.1074/MCP.M600157-MCP200
  150. 150. Liu MC, Akinyi L, Scharf D, Mo J, Larner SF, Muller U, et al. Ubiquitin C-terminal hydrolase-L1 as a biomarker for ischemic and traumatic brain injury in rats. The European Journal of Neuroscience. 2010;31:722-732. DOI: 10.1111/J.1460-9568.2010.07097.X
  151. 151. Papa L, Brophy GM, Welch RD, Lewis LM, Braga CF, Tan CN, et al. Time course and diagnostic accuracy of glial and neuronal blood biomarkers GFAP and UCH-L1 in a large cohort of trauma patients with and without mild traumatic brain injury. JAMA Neurology. 2016;73:551-560. DOI: 10.1001/JAMANEUROL.2016.0039
  152. 152. Bazarian JJ, Biberthaler P, Welch RD, Lewis LM, Barzo P, Bogner-Flatz V, et al. Serum GFAP and UCH-L1 for prediction of absence of intracranial injuries on head CT (ALERT-TBI): A multicentre observational study. Lancet Neurology. 2018;17:782-789. DOI: 10.1016/S1474-4422(18)30231-X
  153. 153. Richard M, Lagares A, Bondanese V, De La Cruz J, Mejan O, Pavlov V, et al. Study protocol for investigating the performance of an automated blood test measuring GFAP and UCH-L1 in a prospective observational cohort of patients with mild traumatic brain injury: European BRAINI study. BMJ Open. 2021;11:e043635. DOI: 10.1136/BMJOPEN-2020-043635
  154. 154. Papa L, Akinyi L, Liu MC, Pineda JA, Tepas JJ, Oli MW, et al. Ubiquitin C-terminal hydrolase is a novel biomarker in humans for severe traumatic brain injury. Critical Care Medicine. 2010;38:138-144. DOI: 10.1097/CCM.0B013E3181B788AB
  155. 155. Mondello S, Linnet A, Buki A, Robicsek S, Gabrielli A, Tepas J, et al. Clinical utility of serum levels of ubiquitin C-terminal hydrolase as a biomarker for severe traumatic brain injury. Neurosurgery. 2012;70:666-675. DOI: 10.1227/neu.0b013e318236a809
  156. 156. Zhang R, Zhang CY, Zhao Q, Li DH. Spectrin: Structure, function and disease. Science China. Life Sciences. 2013;56:1076-1085. DOI: 10.1007/S11427-013-4575-0
  157. 157. Wang KKW, Posmantur R, Nath R, McGinnist K, Whitton M, Talanian RV, et al. Simultaneous degradation of alphaII- and betaII-spectrin by caspase 3 (CPP32) in apoptotic cells. The Journal of Biological Chemistry. 1998;273:22490-22497. DOI: 10.1074/JBC.273.35.22490
  158. 158. Pike BR, Zhao X, Newcomb JK, Posmantur RM, Wang KKW, Hayes RL. Regional calpain and caspase-3 proteolysis of alpha-spectrin after traumatic brain injury. Neuroreport. 1998;9:2437-2442. DOI: 10.1097/00001756-199808030-00002
  159. 159. Pineda JA, Wang KKW, Hayes RL. Biomarkers of proteolytic damage following traumatic brain injury. Brain Pathology. 2004;14:202-209. DOI: 10.1111/J.1750-3639.2004.TB00054.X
  160. 160. Farkas O, Polgár B, Szekeres-Barthó J, Dóczi T, Povlishock JT, Büki A. Spectrin breakdown products in the cerebrospinal fluid in severe head injury--preliminary observations. Acta Neurochirurgica. 2005;147:855-860. DOI: 10.1007/S00701-005-0559-6
  161. 161. Svetlov SI, Larner SF, Kirk DR, Atkinson J, Hayes RL, Wang KKW. Biomarkers of blast-induced neurotrauma: Profiling molecular and cellular mechanisms of blast brain injury. Journal of Neurotrauma. 2009;26:913. DOI: 10.1089/NEU.2008.0609
  162. 162. Li J, Li XY, Feng DF, Pan DC. Biomarkers associated with diffuse traumatic axonal injury: Exploring pathogenesis, early diagnosis, and prognosis. The Journal of Trauma. 2010;69:1610-1618. DOI: 10.1097/TA.0B013E3181F5A9ED
  163. 163. Zetterberg H. Neurofilament light: A dynamic cross-disease fluid biomarker for neurodegeneration. Neuron. 2016;91:1-3. DOI: 10.1016/J.NEURON.2016.06.030
  164. 164. Gaetani L, Blennow K, Calabresi P, Di Filippo M, Parnetti L, Zetterberg H. Neurofilament light chain as a biomarker in neurological disorders. Journal of Neurology, Neurosurgery, and Psychiatry. 2019;90:870-881. DOI: 10.1136/JNNP-2018-320106
  165. 165. Van Geel WJA, Rosengren LE, Verbeek MM. An enzyme immunoassay to quantify neurofilament light chain in cerebrospinal fluid. Journal of Immunological Methods. 2005;296:179-185. DOI: 10.1016/J.JIM.2004.11.015
  166. 166. Laverse E, Guo T, Zimmerman K, Foiani MS, Velani B, Morrow P, et al. Plasma glial fibrillary acidic protein and neurofilament light chain, but not tau, are biomarkers of sports-related mild traumatic brain injury. Brain Communications. 2020;2:fcaa137. DOI: 10.1093/BRAINCOMMS/FCAA137
  167. 167. Shahim P, Politis A, van der Merwe A, Moore B, Ekanayake V, Lippa SM, et al. Time course and diagnostic utility of NfL, tau, GFAP, and UCH-L1 in subacute and chronic TBI. Neurology. 2020;95:e623-e636. DOI: 10.1212/WNL.0000000000009985
  168. 168. Shahim P, Politis A, van der Merwe A, Moore B, Chou YY, Pham DL, et al. Neurofilament light as a biomarker in traumatic brain injury. Neurology. 2020;95:e610-e622. DOI: 10.1212/WNL.0000000000009983
  169. 169. Anderson KJ, Scheff SW, Miller KM, Roberts KN, Gilmer LK, Yang C, et al. The phosphorylated axonal form of the neurofilament subunit NF-H (pNF-H) as a blood biomarker of traumatic brain injury. Journal of Neurotrauma. 2008;25:1079-1085. DOI: 10.1089/NEU.2007.0488

Written By

Vinu V. Gopal, Rinku Raj Mullasseril and Goutam Chandra

Submitted: 09 April 2022 Reviewed: 14 April 2022 Published: 03 June 2022