Next Article in Journal
Acknowledgment to Reviewers of Sports in 2020
Next Article in Special Issue
Determining the Optimal Workrate for Cycle Ergometer Verification Phase Testing in Males with Obesity
Previous Article in Journal
Exercise Intensity during Olympic-Distance Triathlon in Well-Trained Age-Group Athletes: An Observational Study
Previous Article in Special Issue
Applications of the Critical Power Model to Dynamic Constant External Resistance Exercise: A Brief Review of the Critical Load Test
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Factors That Could Affect Concussion Risk in Elite Rugby

1
Sports Genomics Laboratory, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK
2
Sport and Exercise Science, University of Northampton, Northampton NN1 5PH, UK
3
Department of Psychology and Sports Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
4
Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
5
Applied Sports, Technology, Exercise and Medicine (A-STEM) Research Centre, College of Engineering, Swansea University, Swansea SA1 8EN, UK
6
Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
7
Institute of Sport, Exercise and Health, University College London, London WC1E 6BT, UK
*
Author to whom correspondence should be addressed.
Submission received: 15 December 2020 / Revised: 11 January 2021 / Accepted: 18 January 2021 / Published: 22 January 2021

Abstract

:
Elite rugby league and union have some of the highest reported rates of concussion (mild traumatic brain injury) in professional sport due in part to their full-contact high-velocity collision-based nature. Currently, concussions are the most commonly reported match injury during the tackle for both the ball carrier and the tackler (8–28 concussions per 1000 player match hours) and reports exist of reduced cognitive function and long-term health consequences that can end a playing career and produce continued ill health. Concussion is a complex phenotype, influenced by environmental factors and an individual’s genetic predisposition. This article reviews concussion incidence within elite rugby and addresses the biomechanics and pathophysiology of concussion and how genetic predisposition may influence incidence, severity and outcome. Associations have been reported between a variety of genetic variants and traumatic brain injury. However, little effort has been devoted to the study of genetic associations with concussion within elite rugby players. Due to a growing understanding of the molecular characteristics underpinning the pathophysiology of concussion, investigating genetic variation within elite rugby is a viable and worthy proposition. Therefore, we propose from this review that several genetic variants within or near candidate genes of interest, namely APOE, MAPT, IL6R, COMT, SLC6A4, 5-HTTLPR, DRD2, DRD4, ANKK1, BDNF and GRIN2A, warrant further study within elite rugby and other sports involving high-velocity collisions.

1. Introduction

Rugby union (RU) and rugby league (RL) are both full-contact collision-based codes of rugby, which have some of the highest reports of concussion in professional sports (“rugby” will be used to refer to both RU and RL). Rugby-related concussions have been the focus of recent concern over the potential short- and long-term neurodegenerative consequences. In addition, athletes who have had a prior concussion have a higher risk of repeated concussions and subsequent time-loss injury [1,2,3,4]. There is a reported increased risk of potential short- and long-term consequences associated with concussion such as increased injury risk, cognitive impairment, forms of dementia, chronic post-concussion syndrome, migraines, sleep dysfunction, anxiety, post-traumatic stress disorder and second-impact syndrome [5,6,7,8,9,10,11,12,13,14]. These consequences could interrupt or terminate an athletic career, causing short- or long-term ill health.
Sport-related concussion has been defined as a traumatic brain injury (TBI) induced by biomechanical forces [5]. However, many factors contribute to concussion risk such as age, sex, playing position, playing level, behaviour, rules of the sport, neck strength, nutrition, and sleep quality [15,16,17]. Concussion has been widely studied in relation to environmental factors, especially in rugby, where factors considered include activity when concussion occurred (e.g., tackling/being tackled), playing experience, history of concussion, positional differences, use of protective equipment (e.g., headgear/mouth guards) and return-to-play protocols and standard of competition [18,19]. However, a further step to better understanding inter-individual variability involves genetic variation and its association with concussion and related phenotypes. Evidence already exists suggesting an association between several genetic factors and inter-individual variability in traumatic brain injury incidence and severity [20,21,22,23,24,25,26,27,28].
Classical genetic studies (twin or family studies) quantify the heritability of phenotypic traits [27]. As concussion is only experienced by a small proportion of the population [28], recruiting a sufficient number of twins/family members who have experienced concussion is difficult (though not impossible) and has not been undertaken, to our knowledge. Consequently, a classical study on the inheritance of concussion risk, to elucidate the relative contribution of environmental versus genetic factors affecting inter-individual variability in concussion incidence, severity and outcome, would be extremely valuable. Many other sport-related injuries or risk factors for injury have substantial genetic contributions to their inter-individual variability, such as tennis elbow (epicondylitis), for which heritability has been estimated at a substantial ~40% in women [29] and bone mineral density (a predictor of osteoporotic fracture), for which heritability is even greater at 50–85% [30]. Substantial heritability estimates for brain structure (~90%) and cognitive performance (~60%) have also been reported [31,32,33,34]. Given these and other observations of substantial genetic contributions to inter-individual variability in most human traits, it is likely that a substantial genetic component also applies to concussion.
Indeed, the substantial inter-individual variability in injury occurrence, and in outcomes following concussion, is probably due to the interaction of multiple genes in a polygenic manner that reflects the complex pathophysiology [35,36]. Prediction of recovery and future risk is therefore currently difficult [5]. This unexplained inter-individual variability could suggest a future role for genetic screening of concussion-associated risk polymorphisms in order to (i) stratify potential risk of initial injury, for individuals (ii) identify players with a greater risk of prolonged recovery and potential concussion-associated neurological issues, (iii) identify those at risk of repeated concussions, (iv) provide further insight into concussion pathophysiology, and (v) inform concussion management strategies at a practical level in elite sport.
Therefore, the aims of this narrative review are to (1) describe the current data on incidence rates and severity of concussion in elite rugby; (2) provide an overview of the mechanisms and pathophysiology of concussion; (3) evaluate how genetic variation could affect predisposition for and recovery from concussion; and (4) inform the future direction research regarding genetic aspects of concussion in rugby.

2. Incidence Rate and Severity of Concussion in Rugby

The professionalisation of rugby has resulted in alterations in the physical characteristics of players [37,38,39,40]. These alterations in physical characteristics such as body mass, strength, power and speed have increased the physical demands of modern rugby, such as more tackles and rucks per match [40,41,42,43,44]. This increased physicality has contributed to increased incidence rates of concussion in rugby [45,46].
There are many similarities in anthropometric and physiological characteristics of players in RU and RL that reflect comparable physical demands including frequent, heavy physical contact in both rugby codes [40]. Elite rugby (RU and RL) has been reported to have a concussion incidence of ~8–28 concussions per 1000 match hours [47,48], which is lower than sports such as horse racing (17–95) and boxing (13) but higher than sports such as soccer (0.4) [49,50,51]. Seventy percent of head injury assessments in elite RU as a result of a tackle are experienced by the tackler and 30% by the ball carrier [52]. This concussion risk is influenced by athlete speed, playing position, impacting force, body position, type of tackle, tackle technique, and physiological and anthropometric characteristics [53,54].
Recovery from concussion has been defined as a return to sport that encompasses a resolution of post-concussion-related symptoms and a return to clinically normal balance and cognitive functioning [5]. Within 7–10 days, 80–90% of adults with sport-related concussions could be clinically recovered and returned to play (Figure 1) [5,55,56].
For 10–20% of concussion cases, symptoms can persist for >10 days [55]. Time taken to recover from a concussion differs for individuals, as 6.5% of concussed athletes have been reported to not return to play until 14 days post-concussion. For 1.6% of concussed athletes, recovery can take longer than 14 days and these individuals could have chronic post-concussion symptoms for up to 12 months [56,57].
Concussion prevalence during the Rugby World Cups has seen a small increase from ~14% of all injuries in 2015 to ~16% in 2019 [58,59]. In the English Rugby Premiership (the top tier of competition in England), concussion incidence increased dramatically from 8 per 1000 match hours in the 2013–2014 season to 22 in 2016–2017, although this is thought to be largely due to increased awareness and reporting [60]. However, concussion incidence within the English RU Premiership decreased to 18 concussions per 1000 match hours in 2017–2018 (~1 concussion per match) [48]. In elite RL, concussion incidence in the National Rugby League (the top tier of competition in Australia) has ranged from ~9 to 28 concussions per 1000 player match hours over a 17 year period with a tendency to increase over time [61,62,63].
The incidence of concussions in RU is similar for forwards (4–19 per 1000 player match hours) and backs (5–18 per 1000 player match hours) [18,64]. In RL, incidence of concussions ranges from 12 to 48 per 1000 player match hours in forwards and a similar 14 to 44 per 1000 player match hours in backs [65]. Concussion incidence in both codes during training is much lower, accounting for only ~5% of concussions (0.03–0.07 per 1000 player training hours) [18,66]. Fluctuations in incidence over time could be attributed to developments in concussion education or operational strategies such as using ‘Hawkeye’ video analysis [48]. Increased awareness of players, support staff and coaches could account for the increased incidence of concussion reported in recent years [48]. Awareness is thought to be increased due to education initiatives by rugby governing bodies and player associations involving increased recent media attention [67].
The average range of concussion severity in RU ranges from 9 to 21 days absence (period from injury to availability for match selection) [4,48,61,65]. However, inter-individual variability means that severity can range from 2 days to >84 days absence [49]. Data from the 2013–2015 Super League RL seasons suggest severity can range from 9 to 15 days absence [47].

3. Mechanisms of Concussion

Rugby-related concussions can be the result of either direct head contact or inertial causes, but each concussion is a unique event. Contact injuries (e.g., from collisions) cause the brain to impact on the internal surfaces of the skull. Particularly injurious are incidents involving the frontal and temporal fossae regions due to ridges and bony protuberances that deform brain tissue [68]. Kinematic analysis indicates that inertial forces from direct or indirect impacts resulting in angular/linear acceleration/deceleration of the brain from head and neck motions can lead to concussion [69].
The contributions of angular or linear acceleration/deceleration to concussion is debated in the literature [70]. Linear acceleration is associated with changes in pressure gradients within the skull, compared to angular acceleration/deceleration that is associated with shear stresses on the brain forcing tissues to slide over one another and stretch [71]. Shear and stretch mechanical forces stretch axons to the point of axotomy (physical breaking) or partial breaking in areas, such as grey and white matter junctions, small blood vessels and axonal projections [69,72,73].
Concussions appear to vary in impact locations (front, top, back and sides of the head), linear acceleration/deceleration magnitude (61–169 g in collegiate American Football players, although there are concerns about the validity of those high values [74]) and clinical outcomes [75]. However, head impacts from high-magnitude angular acceleration/deceleration result in more severe clinical outcomes due to the propensity of brain tissue to deform more readily from shear forces and are the predominant mechanism in multifocal concussion [71,75]. A tackle or collision may produce whiplash, which in turn produces both linear and angular acceleration/deceleration to the player’s brain [75].

4. Pathophysiology of Concussion

In rugby, the primary mechanical stress injury to neurons is likely the result of a collision that elicits a neuronal stretch. A stretch of ~10–20% of a neuron’s resting length within 100 ms (sublethal axonal injury threshold) can trigger the secondary biochemical response of the neurometabolic cascade [76,77]. The resultant microstructural damage caused by the stretch is hypothesised to be the root cause of all forms of TBI [78,79,80]. The neurometabolic cascade following a concussive event (Figure 2) has been reviewed by Giza and Hovda [76,77].
The initial disturbance and stretch result in the release of depolarising extracellular K+ due to voltage-dependent channels opening in the neuronal membranes and this can last up to 6 h post-concussion [81,82]. Further K+ flux is caused by the release of the excitatory amino acid glutamate [83]. Proteolytic digestion of the axon membrane skeleton occurs due to Ca2+ activation of cysteine proteases and apoptotic genetic signals [84]. Ca2+ influx has been reported to contribute to axonal microtubule breakdown 6–24 h after a concussive event [82]. During smaller insults to the brain, surrounding glial cells remove extracellular K+ in order to maintain homeostasis [85]. However, this cannot be achieved during larger concussive events and greater quantities of excitatory amino acids are released, resulting in ‘spreading depression’ [86]. Multiple mechanisms are responsible for elevated Ca2+ levels—firstly, the physical disruption of membranes through primary injury [87]; secondly, increased glutamate binds receptors such as n-methyl-d aspartic acid (NMDA) subunit NR2A, increasing Ca2+ influx through the NMDA channel, prolonging neuronal dysfunction [88].
Disruption of ionic homeostasis leads to an energy crisis within the injured brain. Re-establishment of ionic homeostasis is further attempted by the employment of ATP-fuelled membrane pumps, which results in increased glycolysis to meet energy requirements due to reduced activity of cerebral oxidative metabolism and reduced cerebral blood flow of up to 50% [89]. Increased intracellular Ca2+, Na+ and K+ can result in swelling and contribute to further reduced cerebral blood flow [90]. Mitochondrial oxidative metabolism is impaired due to the influx of extracellular Ca2+, thus contributing to the energy crisis [91]. As part of the neurometabolic cascade, pro- and anti-inflammatory cytokines are released [92]. Cytokines from this neuroimmune response can play both beneficial and detrimental roles in the neuroinflammatory response following a concussion [92].

5. Genetic Associations with Concussion

Genome-wide association studies (GWAS) enable the genome to be searched for unsuspected variations as opposed to candidate areas as in a gene association study [93,94]. In elite sport, however, the maximum number of individuals available for study is limited. For example, the English Rugby Premiership comprises ~600 players and Super League ~360 players. This limited sample size reduces the feasibility of GWAS, as considerably larger sample sizes are often required to meet the traditionally accepted significance value of p < 5 × 10−8. Genetic association studies utilising a candidate gene approach enable the study of genetic variance within a complex polygenic trait [95]. An advantage of the candidate gene approach is that genes are selected utilising an a priori hypothesis based on the biological function of a particular protein and the specific phenotype [95,96], and statistical power can be sufficient to test specific hypotheses using sample sizes available in elite sport. A disadvantage of the candidate gene approach is that only genes/variants already suspected are investigated, excluding the possibility of discovering hitherto unsuspected genes/variants that might be important.
Functionally, significant polymorphisms (single-nucleotide polymorphisms (SNPs), repeat polymorphisms, insertions or deletions) used in the candidate gene approach are often selected based on the likeliness to affect gene function. Priority polymorphisms include those that alter an amino acid in a protein (missense variation) or produce a stop codon (nonsense variation) [95]. Polymorphisms in promoter and regulatory regions of a gene could also have functional consequences by influencing transcription rate [95].

5.1. Candidate Genetic Variants

A complex array of physiological and psychological responses to concussion have been reported, so the proposed influencing genes have been categorised into four groups. These groups are based on current knowledge and some genes fit into more than one category due to the nature of their functions: 1. genes that affect the severity of concussion; 2. genes that affect repair and plasticity of the brain; 3. genes that affect post-concussion cognitive behavioural capacity; and 4. genes that affect personality traits and concussion risk. The genes are listed in Table 1 and addressed in Section 5.1.1, Section 5.1.2, Section 5.1.3, Section 5.1.4, Section 5.1.5, Section 5.1.6, Section 5.1.7, Section 5.1.8, Section 5.1.9, Section 5.1.10, Section 5.1.11, Section 5.1.12, Section 5.1.13, Section 5.1.14, Section 5.1.15 and Section 5.1.16.

5.1.1. Apolipoprotein E

Apolipoprotein E (APOE) is the most researched gene in respect to TBI. APOE isoforms have both protective and detrimental effects (Supplementary Figure S1). These effects are dependent upon which specific alleles an individual carries and thus gene expression after the TBI event. APOE has three common allelic isoforms ε2, ε3 and ε4 which differ by amino acid substitutions at residues 112 and 158 [148]. Two C/T SNPs at residues 112 (rs429358) and 158 (rs7412) result in amino acid substitutions of arginine (C) to cysteine (T) at each residue (Supplementary Figure S1). The two nonsynonymous SNPs at residues 112 and 158 can produce the three isoforms of ε2, ε3, ε4 and six possible genotypes (Table 2) of relevance to concussion.
APOE isoforms have differing effects on neurite extension, which can influence ability to recover post-concussion. APOE ε3 stimulates neurite growth in cultured neuronal cells [97,98]. In contrast, APOE ε4 suppresses neurite growth [97,98]. These findings suggest that APOE ε2 and ε3 would provide more effective neuronal repair, such as proliferation of dendrites post-concussion compared to APOE ε4 [97,98]. In addition, the ε4 alleles have been associated with the formation of neurodegenerative amyloid plaques (Aβ) and increased risk of Alzheimer’s disease [99].
Despite the pathophysiological roles that APOE ε4 plays in TBI, studies associating APOE ε4 and sport-related concussion are few and findings are conflicting. Kristman et al. [100] showed no association between APOE ε4 carriers and incidence of concussion in Varsity level athletes. These findings have been supported by Terrell et al. [2] and Tierney et al. [1], who also reported no association between concussion incidence and APOE genotypes in collegiate athletes. More recently, Abrahams et al. [149] reported no association in APOE ε2, ε3 and ε4 genotypes and incidence of concussion in a mixed cohort of youth, amateur and professional South African RU players.
Early findings from Jordan et al. [150] indicated that APOE ε4 carrier boxers experiencing high-exposures (>12 professional bouts) had greater chronic brain injury scale scores than non-ε4 carrier high-exposure boxers. Indeed, it has been suggested that the APOE ε4 allele may be responsible for up to 64% of the ‘hazardous influence’ of TBI [151] and athletes who possess the ε4 allele suffer from prolonged physical (Cohen’s d’ = 0.87) and cognitive (d’ = 0.60) symptomatic responses to concussion [152].
Polymorphisms within the promoter region of APOE have been associated with functional regulation of APOE transcription and quantitative impacts on apolipoprotein E levels in brain tissue, as well as unfavourable outcomes post-TBI [101,102]. It has been hypothesised that the -219 T allele at rs405509 exacerbates the effects of the ε4 allele through upregulation of APOE gene transcription and increased Aβ plaque accumulation [102].
Lendon et al. [102] observed an association between individuals with rs405509 TT genotype and unfavourable outcomes post-TBI over a six-month recovery period. Tierney et al. [1] reported that carriers of the T allele had an 8-fold greater risk of experiencing two or more concussions. Similarly, Terrell et al. [2] suggest that the TT genotype is associated with a 3-fold greater risk of previous concussion and a 4-fold greater risk of a history of concussion with loss of consciousness. In contrast, Abrahams et al. [149] reported that TT genotype was associated with a 45% reduced risk of and concussion and the T allele was associated with a <1-week recovery period post-concussion in a mixed cohort of youth and professional South African RU players. These conflicting findings could be in part due to differences in sport and, in particular, geographic ancestry of the participants. Nevertheless, the plausible physiological mechanisms and the limited number of association studies warrant further investigation of this concussion-associated SNP.

5.1.2. Microtubule-Associated Protein Tau Polymorphisms

The functions of microtubule-associated protein tau (MAPT) include encoding the tau protein that modulates microtubule formation, structural stabilisation of the neuronal axons and driving growth of neurites [103,104]. Elevated post-TBI plasma levels of tau have been observed for up to 90 days [153]. Autopsies on American football players’ brains who had experienced repetitive concussions indicate the presence of neurofibrillary tangles (aggregates of hyperphosphorylated tau protein) and neuropil filaments (abnormal neurite formations) [154]. These neurotoxic formations have been associated with neurodegenerative diseases such as Alzheimer’s disease, chronic traumatic encephalopathy, Parkinson’s disease, frontotemporal dementia and a range of other neurodegenerative diseases under the term tauopathies [155,156,157,158]. The MAPT (rs10445337) T/C SNP is postulated to modulate the formation of neurotoxic-paired helical filaments composed of hyperphosphorylated tau [159,160] (Supplementary Figure S2).
Terrell et al. [2] reported a nonsignificant observation that the MAPT rs10445337 TT genotype was weakly associated with a history of one or more concussions (odds ratio, 2.1; 95% CI, 0.3 to 14.5). Similarly, in a later study, no association was observed between concussion incidence and MAPT rs10445337 [22]. Recently, other MAPT SNPs (rs2435211 and rs2435200) have been implicated as potential pathophysiological mechanisms in RU players [20]. The AG genotype of rs2435200 has been associated with an increased risk of sustaining multiple concussions in senior (>18 years old) RU players [20]. In addition, the T-G haplotype (rs2435211 and rs2435200) has been associated with an increased risk of sustaining a concussion in senior amateur and elite RU players [20].

5.1.3. Neurofilament Heavy Polymorphism

Approximately 50% of the neuronal cytoskeleton is comprised of light, medium and heavy neurofilaments [105]. A function of the neuronal cytoskeleton is to resist the resultant strain caused by biomechanical forces during a head impact [105]. In one study, a small cohort of 48 college level athletes with self-reported history of concussion were genotyped for an A/C polymorphism (rs165602) of the neurofilament heavy (NEFH) gene (Supplementary Figure S3) [161]. The authors observed no association between the polymorphism and incidence or severity of concussion in college athletes.

5.1.4. Membrane Metalloendopeptidase Polymorphism

The membrane metalloendopeptidase (MME) gene encodes the neprilysin protease (Supplementary Figure S4) that degrades amyloid plaque (Aβ) proteins [106]. A GT repeat within the promoter region of MME regulates expression of neprilysin in neurons [107]. Greater Aβ deposits were observed after severe TBI in patients who had long MME GT repeats (>41) [108]. It was also observed that carrying at least one 22-repeat allele was associated with increased risk of Aβ plaque deposition and carrying at least one 20-repeat allele associated with decreased risk.

5.1.5. Brain-Derived Neurotrophic Factor Polymorphism

Brain-derived neurotrophic factor (BDNF) is a gene that affects the repair and plasticity of neurons. It is a member of the neurotrophin family, responsible for mediating neuronal plasticity [109,110]. Neurotrophins aid in the development, differentiation, proliferation and survival of neurons (dopaminergic, serotonergic and cholinergic) [109,111]. A widely studied SNP is the C to T missense variation at nucleotide 196 resulting in a valine to methionine (Val66Met) substitution at codon 66 [162] (Supplementary Figure S5). BDNF mRNA is upregulated post-TBI event and can remain elevated for up to three days post-TBI [162,163,164,165]. BDNF plays an important role in strengthening existing synaptic connections and modulating the creation of new synapses [110]. The Met allele impairs intracellular tracking and packaging of precursor-BDNF (pro-BDNF) and activity-dependent secretion of BDNF [162].
Dretsch et al. [166] reported that ~17% of Met/Met homozygotes suffered a concussion during military deployment compared to ~4% of Val carriers. Narayanan et al. [167] found that the rs6265 polymorphism was associated with neurocognitive performance in concussed individuals acutely and 6 months post-event, as Val/Val homozygotes performed better in measures of memory, executive function, attention and overall cognitive performance [167].

5.1.6. Glutamate Ionotropic Receptor NMDA Type Subunit 2A Variant

Glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A) encodes glutamate-gated ion channel proteins. A variable number tandem repeat (VNTR) polymorphism within the promoter region of GRIN2A modulates n-methyl-d aspartic acid (NMDA) receptors within the brain. The NMDA NR2A subunit has been associated with neuronal plasticity, spatial and episodic memory [112,113]. The VNTR GT (rs3219790) repeat within the promoter region affects transcriptional activity in a length-dependent manner (Supplementary Figure S6) [114,115]. The longer the GT repeat, the lower the GRIN2A promoter activity [115]. Longer repeats of >25 (GT) can be termed long alleles (L) and shorter repeats of <25 (GT) termed short alleles (S) [114,115].
Findings from McDevitt et al. [168] indicate that L allele carriers were twice as likely to recover in >60 days than S allele carriers. A dose response was also reported: LL carriers were 6-fold more likely to have a prolonged recovery (>60 days) compared to individuals of SS genotype.

5.1.7. Catechol-O-methyltransferase Polymorphism

The catechol-O-methyltransferase (COMT) gene has been postulated to affect post-concussion cognitive behavioural capacity [116]. COMT encodes an enzyme that methylates and in turn deactivates catechol-based neurotransmitters such as synaptic dopamine and noradrenaline [117] (Supplementary Figure S7). Optimal cognitive function is affected by the prefrontal cortex’s sensitivity to dopamine, which makes COMT an ideal candidate gene for influencing inter-individual variability in cognitive function post-concussion. A widely studied SNP within the COMT gene is the G to A missense variation at codon 158 resulting in a valine (Val) to methionine (Met) amino acid substitution. Val/Val carriers have greater COMT activity than Met/Met carriers [118].
Lipsky et al. [116] reported that Val allele carriers performed poorer on tests of executive function compared to Met allele carriers post-TBI. More recently and in contrast, Willmott et al. [169] reported no significant influence of COMT polymorphisms on cognitive performance in moderate to severe TBI patients. However, Lipsky et al. [116] employed a battery of executive function tests including the Wisconsin Card Sorting Test, while Willmott et al. [169] used the Glasgow Outcome Scale-Extended as a measure of functional outcome post-TBI. Mc Fie et al. [21] reported that Met carriers in a cohort of youth and professional South African RU players were ~3-fold more likely to have a history of concussion and, accordingly, it has been postulated that elevated dopamine could increase impulsivity and risk taking meaning Met allele carriers could place themselves at increased risk of sustaining a concussion [170,171].

5.1.8. Ankyrin Repeat and Kinase Domain Containing 1 Polymorphism

Ankyrin repeat and kinase domain containing 1 (ANKK1) is a dopaminergic gene known to affect working memory, reward and motivation [119,120]. ANKK1 was originally referred to as Taq1A and is in linkage disequilibrium (D’ > 0.80) with the 10 kB downstream dopamine receptor D2 (DRD2) gene [121]. The ANKK1 C/T (rs1800497) SNP is hypothesised to be in a regulatory region within DRD2 (Supplementary Figure S8) [121]. ANKK1 is expressed in astroglial cells (a type of brain-derived glial cell), post-mitotic neurons and neural precursors from neurogenic niches and as a member of the serine/threonine receptor-interacting protein kinases is responsible for dopaminergic signal transduction and cellular response [121,122].
ANKK1 polymorphisms affect dopamine transporter densities within the striatum which influences working memory, reward and motivation [121,122]. The T allele of ANKK1 has been associated with a 30–40% reduction in the expression of D2 receptors within the ventral striatum [123,124]. ANKK1’s polymorphic role in modulating working memory and cognitive performance vis-à-vis concussion/TBI is limited to three studies. McAllister et al. [172,173] observed concussed T allele carriers performed significantly worse in measures of learning, working memory and response latencies. Similarly, Yue et al.’s [174] findings support McAllister et al. [172,173] and indicate a dose-dependent association with the T allele. Thus, this polymorphism could influence recovery from a concussive event.

5.1.9. Dopamine Receptor-Related Polymorphisms

Dopamine receptors (DRD2 and DRD4) have been associated with risk-taking behaviours (impulsivity, behavioural inhibition and novelty seeking) [125,126]. Polymorphisms within DRD2 and DRD4 genes have been postulated to affect personality traits, possibly via inhibition of neurotransmission [175]. DRD2 SNPs rs12364283 (A/G) and rs1076560 (C/A) have been associated with altered D2 receptor expression (Supplementary Figure S9) [175]. The DRD4 promoter rs1800955 C allele has been associated with higher DRD4 expression compared to the T allele (Supplementary Figure S9) [125]. Furthermore, the DRD4 (rs1800955) CC genotype and inferred haplotype of DRD2 (rs12364283–rs1076560)–DRD4 (rs1800955) A–C–C alleles associated with decreased concussion susceptibility in junior South African RU players (12–18 years old) [176]. It is suggested that carriers of the DRD4 (rs1800955) C allele could have reduced concussion susceptibility via a neuro-protective response from greater D4 receptor availability, thus inhibiting risk-taking behaviour.

5.1.10. Serotonin Transporter Polymorphisms

The serotonin transporter gene (solute carrier family 6 member 4, SLC6A4) is reported to play a role in personality and behavioural traits [127]. The 5-HTTLPR (rs4795541) polymorphism is a variable number tandem repeat (up to 28 bp) insertion (long (L) allele) or deletion (short (S) allele) located in the promoter region of the 5-HTT-encoding gene SLC6A4. Reduced serotonin transporter expression is reported for the S allele (Supplementary Figure S10) [128]. An additional an A/G SNP (rs25531) within the long allele of rs4795541 appears to modulate serotonin transporter expression further, as the LG allele has been associated with lower serotonin expression than the LA allele [177]. The S allele of rs4795541 has previously been associated with harm avoidance, impulsive behaviours and risk taking, though inconsistently. In 78 sibling pairs, harm avoidance scores were higher for S allele carriers than L allele carriers [129] and individuals possessing the LL genotype have been observed to be more risk taking during decision-making trials [130]. However, children and adolescents carrying the S allele showed more impulsive behaviour such as delay aversion during target-game activity [131]. Recently, it has been observed that 5-HTTLPR low (SA/SA) and intermediate (SA/LA, SA/LG, LA/LG, LG/LG)-possessing junior RU players displayed less harm avoidance behaviour [21]. These findings suggest that genetic variants associated with personality and thus behavioural traits could influence concussion risk in rugby.

5.1.11. Endothelial Nitric Oxide Synthase Polymorphism

Nitric oxide (NO) plays a major role in the maintenance of cerebral blood flow and is synthesised by three NO synthase isoforms—endothelial (eNOS), neuronal and inducible [132,133]. Nitric oxide is reduced post-TBI under experimental conditions [134,135] and the NOS3 -786T/C (rs20707044) promoter polymorphism has been associated with promoter region activity, reduced NO synthesis and cerebral vasospasm [136] (Supplementary Figure S11).
Robertson et al. [137] reported lower cerebral blood flow in -786 C allele (rs2070744)-carrying patients with severe TBI. Multifactorial pathophysiological mechanisms contribute to the reduction in cerebral blood flow as a result of sustaining a concussion [89]. Thus, it could be postulated that possession of a -786 C allele could negatively affect a concussed individual, due to further reduced cerebral blood flow and this warrants further investigation.

5.1.12. Angiotensin I-Converting Enzyme Variants

Cerebral blood flow and autoregulation can be reduced following TBI [178]. The angiotensin I-converting enzyme (ACE) (rs4646994) insertion (I)/deletion (D) polymorphism (Supplementary Figure S12) has been associated with regulating blood pressure and cerebral circulation [138]. The DD genotype is associated with higher ACE activity [138] and the D allele has been associated with worse cognitive and motor outcome one month after moderate–severe TBI [139]. Other ACE polymorphisms (rs7221780 and rs8066276) have been associated with worse Glasgow Outcome Scale scores 6 months post-TBI [140].

5.1.13. Tumour Necrosis Factor Polymorphisms

Inflammatory mediator cytokines can play contrasting roles in TBI, as they could exacerbate effects in early phases and could affect recovery and repair in the later phases [87,179]. Immediately post-TBI, proinflammatory cytokine tumour necrosis factors (TNFs) are upregulated and return to baseline levels within 24-h [180]. TNFs mediate neuronal apoptosis in the early phase of TBI and facilitate repair in the long term [141,142]. In patients with moderate–severe TBI, carriers of an A allele at position TNF-308 (rs1800629) (Supplementary Figure S13) had an increased risk of unfavourable outcome six months post-TBI compared to noncarriers [143]. Located in the promoter region of TNF, the A allele has been associated with increased gene expression and as a result is postulated to increase risk of unfavourable outcome post-TBI [143,181].

5.1.14. Transforming Growth Factor Beta1 Polymorphism

The suppressive cytokine transforming growth factor beta1 (TGFB1) plays a role in regulating inflammation and is encoded by the transforming growth factor beta 1 (TGFB1) gene [144]. Two polymorphisms within the promoter region of TGFB1 (-800 G/A rs1800468 and -509 C/T rs1800469) (Supplementary Figure S14) have been associated with altered TGFB plasma levels [145]. However, Waters et al. [143] reported no association between these TGFB1 polymorphisms and overall outcome in severe TBI patients.

5.1.15. Interleukin 1 Alpha and Interleukin 1 Beta Polymorphisms

Interleukin 1 alpha (IL1A) and interleukin 1 beta (IL1B) are proinflammatory cytokines (Supplementary Figure S15). In experimental models, both IL1A and IL1B levels are increased within hours following a TBI and can remain elevated for days [182]. There are inconsistent findings regarding IL1A and IL1B polymorphisms and outcome post-TBI. The G allele of IL1B -511 (rs16944) and the T allele of +3953 (rs1143634) have been associated with a six-month unfavourable outcome in severe TBI patients [146]. However, Waters et al. [143] observed no association between IL1A and IL1B polymorphisms and a six-month unfavourable outcome. Furthermore, associations with secondary complications such as seizures and raised intracranial pressure have been reported for the T allele of IL1A -899 (rs1800587) and the T allele of IL1B +3953 (rs1143634).

5.1.16. Interleukin 6 Receptor Polymorphism

Interleukin 6 plays a role in the inflammatory process following injury through both pro- and anti-inflammatory properties [147]. A SNP exists at residue 358 (rs2228145) of the interleukin 6 receptor (IL6R) gene (Supplementary Figure S16), the CC genotype of which has been associated with an increased risk of concussion in college athletes [22]. It is postulated that the CC genotype could increase the early inflammatory response post-concussion and lead to reduced cognition [22]. However, Waters et al. [143] reported no associations between IL6R promotor polymorphisms and outcome in severe-TBI patients.

6. Conclusions and Future Directions

Elite rugby players are exposed to a higher risk of concussion during a playing career than athletes in many other sports. A critical step in better understanding inter-individual variability in the risk of sustaining a concussion and the duration of recovery following a concussion involves identifying genetic variations associated with those risks. The literature has already identified several genetic factors with inter-individual variability in concussion and TBI incidence, severity and recovery. The genes and polymorphisms reviewed here, along with many others, need to be investigated further in relation to incidence rates and recovery from concussion, particularly in a sport such as rugby with a relatively high concussion risk. The number of individuals competing in truly elite rugby is low, so highly collaborative research is required to achieve sample sizes sufficient for satisfactory statistical power.
The inter-individual variation in outcomes following concussion makes predictions of recovery and future risk difficult. This variability could mean there is a future valuable role for genetic screening of concussion-associated risk polymorphisms to complement other data. Achieving elite status in a sport such as rugby is a multifactorial accomplishment due to the complex interactions of multiple environmental factors and the polygenic nature of inherited characteristics and predispositions. Epigenetic regulation of genome function in the context of particular environmental stimuli might also be important in modulating the risk of concussion injury and the rate of recovery. Elite rugby players are exposed to one of the highest risks of concussion in team sports, so distinctive genetic characteristics may exist in those athletes that offer advantages in resisting frequent or severe concussions, relative to those less successful in the sport. Athletes in other sports with a high risk of concussion are also particularly likely to benefit from this kind of genetic resistance to injury. The findings, however, could be applied to a wider range of sports, including those with a lower but still extant risk of concussion. Thus, future research that combines an individual’s concussion history and other phenotypes with detailed genomic information could facilitate more personalised management of concussion and eventually help protect athletes from unfavourable longer-term health outcomes.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/2075-4663/9/2/19/s1, Figure S1: Schematic of APOE variants, Figure S2: Schematic of MAPT polymorphisms, Figure S3: Schematic of rs165602 NEFH, Figure S4: Schematic of GT repeat promoter polymorphism located in MME gene, Figure S5: Schematic of BDNF (rs6265), Figure S6: Schematic of GRIN2A promoter (rs3219790), Figure S7: Schematic of rs4680 Catechol-O-methyltransferase, Figure S8: Schematic of ankyrin repeat and kinase domain containing 1 (rs1800497), Figure S9: Schematic of DRD2 (rs1076560 and rs12364283) and DRD4 (rs1800955), Figure S10: Schematic of rs4795541 5-HTTLPR, in conjunction with rs25531, Figure S11: Schematic of rs2070744 NOS3, Figure S12: Schematic of ACE (rs4646994), Figure S13: Schematic of TNF (rs1800629), Figure S14: Schematic of Transforming growth factor beta1 (rs1800468 and rs1800469), Figure S15: Schematic of Interleukin 1 alpha (rs1800587) and Interleukin 1 beta (rs16944 and rs1143634), Figure S16: Schematic of rs2228145 interleukin 6 receptor.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article or Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tierney, R.; Mansell, J.; Higgins, M.; McDevitt, J.; Toone, N.; Gaughan, J.; Mishra, A.; Krynetskiy, E. Apolipoprotein E Genotype and Concussion in College Athletes. Clin. J. Sport Med. 2010, 20, 464–468. [Google Scholar] [CrossRef] [PubMed]
  2. Terrell, T.; Bostick, R.; Abramson, R.; Xie, D.; Barfield, W.; Cantu, R.; Stanek, M.; Ewing, T. APOE, APOE Promoter, and Tau Genotypes and Risk for Concussion in College Athletes. Clin. J. Sport Med. 2008, 18, 10–17. [Google Scholar] [CrossRef] [PubMed]
  3. Terrell, T.; Bostick, R.; Barth, J.; McKeag, D.; Cantu, R.; Sloane, R.; Galloway, L.; Erlanger, D.; Valentine, V.; Bielak, K. Genetic Polymorphisms, Concussion Risk, and Post Concussion Neurocognitive Deficits in College and High School Athletes. Br. J. Sports Med. 2013, 47, e1. [Google Scholar] [CrossRef]
  4. Cross, M.; Kemp, S.; Smith, A.; Trewartha, G.; Stokes, K. Professional Rugby Union Players Have a 60% Greater Risk of Time Loss Injury after Concussion: A 2-Season Prospective Study of Clinical Outcomes. Br. J. Sports Med. 2016, 50, 926–931. [Google Scholar] [CrossRef] [Green Version]
  5. McCrory, P.; Meeuwisse, W.; Dvorak, J.; Aubry, M.; Bailes, J.; Broglio, S.; Cantu, B.; Cassidy, D.; Echemendia, R.J.; Castellani, R.J.; et al. Consensus Statement on Concussion in Sport—the 5th International Conference on Concussion in Sport Held in Berlin, October 2016. Br. J. Sports Med. 2018, 51, 838–847. [Google Scholar]
  6. Cunningham, J.; Broglio, S.; Wilson, F. Influence of Playing Rugby on Long-Term Brain Health Following Retirement: A Systematic Review and Narrative Synthesis. BMJ Open Sport Exerc. Med. 2018, 4, e000356. [Google Scholar] [CrossRef]
  7. Blennow, K.; de Leon, M.; Zetterberg, H. Alzheimer’s Disease. Lancet 2006, 368, 387–403. [Google Scholar] [CrossRef]
  8. Hume, P.; Theadom, A.; Lewis, G.; Quarrie, K.; Brown, S.; Hill, R.; Marshall, S. A Comparison of Cognitive Function in Former Rugby Union Players Compared with Former Non-Contact-Sport Players and the Impact of Concussion History. Sports Med. 2017, 47, 1209–1220. [Google Scholar] [CrossRef]
  9. Quintana, L.M. Second Impact Syndrome in Sports. World Neurosurg. 2016, 91, 647–649. [Google Scholar] [CrossRef]
  10. Broglio, S.P.; Eckner, J.T.; Paulson, H.L.; Kutcher, J. Cognitive Decline and Aging. Exerc. Sport Sci. Rev. 2012, 40, 138–144. [Google Scholar] [CrossRef]
  11. Lee, Y.-K.; Hou, S.-W.; Lee, C.-C.; Hsu, C.-Y.; Huang, Y.-S.; Su, Y.-C. Increased Risk of Dementia in Patients with Mild Traumatic Brain Injury: A Nationwide Cohort Study. PLoS ONE 2013, 8, e62422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kerr, Z.Y.; Evenson, K.R.; Rosamond, W.D.; Mihalik, J.P.; Guskiewicz, K.M.; Marshall, S.W. Association between Concussion and Mental Health in Former Collegiate Athletes. Inj. Epidemiol. 2014, 1, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Guskiewicz, K.M.; Marshall, S.W.; Bailes, J.; McCrea, M.; Harding, H.P.; Matthews, A.; Mihalik, J.R.; Cantu, R.C. Recurrent Concussion and Risk of Depression in Retired Professional Football Players. Med. Sci. Sport. Exerc. 2007, 39, 903–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Stulemeijer, M.; Andriessen, T.M.; Brauer, J.M.P.; Vos, P.E.; Van Der Werf, S. Cognitive Performance after Mild Traumatic Brain Injury: The Impact of Poor Effort on Test Results and Its Relation to Distress, Personality and Litigation. Brain Inj. 2007, 21, 309–318. [Google Scholar] [CrossRef] [PubMed]
  15. Raikes, A.C.; Athey, A.; Alfonso-Miller, P.; Killgore, W.D.S.; Grandner, M.A. Insomnia and Daytime Sleepiness: Risk Factors for Sports-Related Concussion. Sleep Med. 2019, 58, 66–74. [Google Scholar] [CrossRef] [Green Version]
  16. Lust, C.A.C.; Mountjoy, M.; Robinson, L.E.; Oliver, J.M.; Ma, D.W.L. Sports-Related Concussions and Subconcussive Impacts in Athletes: Incidence, Diagnosis, and the Emerging Role of EPA and DHA. Appl. Physiol. Nutr. Metab. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Abrahams, S.; Mc Fie, S.; Patricios, J.; Posthumus, M.; September, A.V. Risk Factors for Sports Concussion: An Evidence-Based Systematic Review. Br. J. Sports Med. 2013. [Google Scholar] [CrossRef]
  18. Gardner, A.J.; Iverson, G.L.; Williams, W.H.; Baker, S.; Stanwell, P. A Systematic Review and Meta-Analysis of Concussion in Rugby Union. Sports Med. 2014, 44, 1717–1731. [Google Scholar] [CrossRef]
  19. Gardner, A.; Iverson, G.L.; Levi, C.R.; Schofield, P.W.; Kay-Lambkin, F.; Kohler, R.M.N.; Stanwell, P. A Systematic Review of Concussion in Rugby League. Br. J. Sports Med. 2015, 49, 495–498. [Google Scholar] [CrossRef] [Green Version]
  20. Abrahams, S.; Mc Fie, S.; Patricios, J.; Suter, J.; September, A.V.; Posthumus, M. Toxic Tau: The TAU Gene Polymorphisms Associate with Concussion History in Rugby Union Players. J. Sci. Med. Sport 2019, 22, 22–28. [Google Scholar] [CrossRef]
  21. Mc Fie, S.; Abrahams, S.; Patricios, J.; Suter, J.; Posthumus, M.; September, A.V. The Association between COMT Rs4680 and 5-HTTLPR Genotypes and Concussion History in South African Rugby Union Players. J. Sports Sci. 2018, 36, 920–933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Terrell, T.R.; Abramson, R.; Barth, J.T.; Bennett, E.; Cantu, R.C.; Sloane, R.; Laskowitz, D.T.; Erlanger, D.M.; McKeag, D.; Nichols, G.; et al. Genetic Polymorphisms Associated with the Risk of Concussion in 1056 College Athletes: A Multicentre Prospective Cohort Study. Br. J. Sports Med. 2018, 52, 192–198. [Google Scholar] [CrossRef] [PubMed]
  23. Davidson, J.; Cusimano, M.D.; Bendena, W.G. Post-Traumatic Brain Injury. Neuroscience 2015, 21, 424–441. [Google Scholar] [CrossRef] [PubMed]
  24. Weaver, S.M.; Portelli, J.N.; Chau, A.; Cristofori, I.; Moretti, L.; Grafman, J. Genetic Polymorphisms and Traumatic Brain Injury: The Contribution of Individual Differences to Recovery. Brain Imaging Behav. 2014, 8, 420–434. [Google Scholar] [CrossRef]
  25. McAllister, T.W. Genetic Factors Modulating Outcome after Neurotrauma. PM&R 2010, 2 (Suppl. 2), S241–S252. [Google Scholar] [CrossRef]
  26. Wilson, M.; Montgomery, H. Impact of Genetic Factors on Outcome from Brain Injury. Br. J. Anaesth. 2007, 99, 43–48. [Google Scholar] [CrossRef] [Green Version]
  27. Mayhew, A.J.; Meyre, D. Assessing the Heritability of Complex Traits in Humans: Methodological Challenges and Opportunities. Curr. Genom. 2017, 18, 332. [Google Scholar] [CrossRef] [Green Version]
  28. Kazl, C.; Torres, A. Definition, Classification, and Epidemiology of Concussion. Semin. Pediatr. Neurol. 2019, 30, 9–13. [Google Scholar] [CrossRef]
  29. Hakim, A.J.; Cherkas, L.F.; Spector, T.D.; MacGregor, A.J. Genetic Associations between Frozen Shoulder and Tennis Elbow: A Female Twin Study. Rheumatology 2003, 42, 739–742. [Google Scholar]
  30. Ralston, S.H.; Uitterlinden, A.G. Genetics of Osteoporosis. Endocr. Rev. 2010, 31, 629–662. [Google Scholar] [CrossRef] [Green Version]
  31. Carmelli, D.; DeCarli, C.; Swan, G.E.; Jack, L.M.; Reed, T.; Wolf, P.A.; Miller, B.L. Evidence for Genetic Variance in White Matter Hyperintensity Volume in Normal Elderly Male Twins. Stroke 1998, 29, 1177–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Geschwind, D.H.; Miller, B.L.; DeCarli, C.; Carmelli, D. Heritability of Lobar Brain Volumes in Twins Supports Genetic Models of Cerebral Laterality and Handedness. Proc. Natl. Acad. Sci. USA 2002, 99, 3176–3181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Carmelli, D.; Swan, G.E.; DeCarli, C.; Reed, T. Quantitative Genetic Modeling of Regional Brain Volumes and Cognitive Performance in Older Male Twins. Biol. Psychol. 2002, 61, 139–155. [Google Scholar] [CrossRef]
  34. Bartley, A.J.; Jones, D.W.; Weinberger, D.R. Genetic Variability of Human Brain Size and Cortical Gyral Patterns. Brain 1997, 120, 257–269. [Google Scholar] [CrossRef]
  35. McKee, A.C.; Daneshvar, D.H.; Alvarez, V.E.; Stein, T.D. The Neuropathology of Sport. Acta Neuropathol. 2014, 127, 29–51. [Google Scholar] [CrossRef]
  36. Dashnaw, M.L.; Petraglia, A.L.; Bailes, J.E. An Overview of the Basic Science of Concussion and Subconcussion: Where We Are and Where We Are Going. Neurosurg. Focus 2012, 33, E5. [Google Scholar] [CrossRef] [Green Version]
  37. Sedeaud, A.; Marc, A.; Schipman, J.; Tafflet, M.; Hager, J.-P.; Toussaint, J.-F. How They Won Rugby World Cup through Height, Mass and Collective Experience. Br. J. Sports Med. 2012, 46, 580–584. [Google Scholar] [CrossRef]
  38. Austin, D.; Gabbett, T.; Jenkins, D. The Physical Demands of Super 14 Rugby Union. J. Sci. Med. Sport 2011, 14, 259–263. [Google Scholar] [CrossRef]
  39. Duthie, G.; Pyne, D.; Hooper, S. Applied Physiology and Game Analysis of Rugby Union. Sports Med. 2003, 33, 973–991. [Google Scholar] [CrossRef]
  40. Brazier, J.; Antrobus, M.; Stebbings, G.K.; Day, S.H.; Callus, P.; Erskine, R.M.; Bennett, M.A.; Kilduff, L.P.; Williams, A.G. Anthropometric and Physiological Characteristics of Elite Male Rugby Athletes. J. Strength Cond. Res. 2020, 34, 1790–1801. [Google Scholar] [CrossRef]
  41. Hill, N.; Rilstone, S.; Stacey, M.; Amiras, D.; Chew, S.; Flatman, D.; Oliver, N. Changes in Northern Hemisphere Male International Rugby Union Players’ Body Mass and Height between 1955 and 2015. BMJ Open Sport Exerc. Med. 2018, 4, e000459. [Google Scholar] [CrossRef] [PubMed]
  42. Johnston, R.D.; Gabbett, T.J.; Jenkins, D.G. Applied Sport Science of Rugby League. Sports Med. 2014, 44, 1087–1100. [Google Scholar] [CrossRef] [PubMed]
  43. Quarrie, K.L.; Hopkins, W.G. Changes in Player Characteristics and Match Activities in Bledisloe Cup Rugby Union from 1972 to 2004. J. Sports Sci. 2007, 25, 895–903. [Google Scholar] [CrossRef] [PubMed]
  44. Eaves, S.; Hughes, M. Patterns of Play of International Rugby Union Teams before and after the Introduction of Professional Status. Int. J. Perform. Anal. Sport 2003, 3, 103–111. [Google Scholar] [CrossRef]
  45. Williams, S.; Trewartha, G.; Kemp, S.; Stokes, K. A Meta-Analysis of Injuries in Senior Men’s Professional Rugby Union. Sports Med. 2013, 43, 1043–1055. [Google Scholar] [CrossRef] [Green Version]
  46. King, D.A.; Hume, P.A.; Milburn, P.D.; Guttenbeil, D. Match and Training Injuries in Rugby League. Sports Med. 2010, 40, 163–178. [Google Scholar] [CrossRef]
  47. Fitzpatrick, A.; Naylor, A.; Myler, P.; Robertson, C. A Three-Year Epidemiological Prospective Cohort Study of Rugby League Match Injuries from the European Super League. J. Sci. Med. Sport 2018, 21, 160–165. [Google Scholar] [CrossRef]
  48. England Professional Rugby Injury Surveillance Project Steering Group. England Professional Rugby Injury Surveillance Project 2017–2018 Season Report; England Professional Rugby Injury Surveillance Project Steering Group: London, UK, 2019; p. 15. [Google Scholar]
  49. Clay, M.B.; Glover, K.L.; Lowe, D.T. Epidemiology of Concussion in Sport: A Literature Review. J. Chiropr. Med. 2013, 12, 230–251. [Google Scholar] [CrossRef] [Green Version]
  50. Koh, J.O.; Cassidy, J.D.; Watkinson, E.J. Incidence of Concussion in Contact Sports: A Systematic Review of the Evidence. Brain Inj. 2003, 17, 901–917. [Google Scholar] [CrossRef]
  51. Chachad, S.; Khan, A. Concussion in the Athlete: A Review. Clin. Pediatr. 2006, 285–288. [Google Scholar] [CrossRef]
  52. Tucker, R.; Raftery, M.; Fuller, G.; Hester, B.; Kemp, S.; Cross, M. A Video Analysis of Head Injuries Satisfying the Criteria for a Head Injury Assessment in Professional Rugby Union: A Prospective Cohort Study. Br. J. Sports Med. 2017, 51, 1147–1151. [Google Scholar] [CrossRef] [PubMed]
  53. Gabbett, T.J.; Jenkins, D.G.; Abernethy, B. Physical Collisions and Injury in Professional Rugby League Match-Play. J. Sci. Med. Sport 2011, 14, 210–215. [Google Scholar] [CrossRef] [PubMed]
  54. Fuller, C.W.; Ashton, T.; Brooks, J.H.M.; Cancea, R.J.; Hall, J.; Kemp, S.P.T. Injury Risks Associated with Tackling in Rugby Union. Br. J. Sports Med. 2010, 44, 159–167. [Google Scholar] [CrossRef] [PubMed]
  55. McCrory, P.; Meeuwisse, W.H.; Aubry, M.; Cantu, R.C.; Dvořák, J.; Echemendia, R.J.; Engebretsen, L.; Johnston, K.; Kutcher, J.S.; Raftery, M.; et al. Consensus Statement on Concussion in Sport: The 4th International Conference on Concussion in Sport, Zurich, November 2012. J. Athl. Train. 2013, 48, 554–575. [Google Scholar] [CrossRef] [Green Version]
  56. Iverson, G.L.; Brooks, B.L.; Collins, M.W.; Lovell, M.R. Tracking Neuropsychological Recovery Following Concussion in Sport. Brain Inj. 2006, 20, 245–252. [Google Scholar] [CrossRef]
  57. Pellman, E.J.; Viano, D.C.; Casson, I.R.; Arfken, C.; Powell, J. Concussion in Professional Football: Injuries Involving 7 or More Days out—Part 5. Neurosurgery 2004, 55, 1100–1116. [Google Scholar] [CrossRef]
  58. Fuller, C.W.; Taylor, A.; Kemp, S.P.T.; Raftery, M. Rugby World Cup 2015: World Rugby Injury Surveillance Study. Br. J. Sports Med. 2017, 51, 51–57. [Google Scholar] [CrossRef] [Green Version]
  59. Fuller, C.W.; Taylor, A.; Douglas, M.; Rafter, M.; Taylor, A.; Douglas, M.; Raftery, M. Rugby World Cup 2019 Injury Surveillance Study. S. Afr. J. Sports Med. 2020, 32, 1–6. [Google Scholar] [CrossRef]
  60. Rafferty, J.; Ranson, C.; Oatley, G.; Mostafa, M.; Mathema, P.; Crick, T.; Moore, I.S. On Average, a Professional Rugby Union Player Is More Likely than Not to Sustain a Concussion after 25 Matches. Br. J. Sports Med. 2019, 53, 969–973. [Google Scholar] [CrossRef] [Green Version]
  61. Video Review and Validation Study. Sports Med. Open 2017, 3, 29. [CrossRef] [Green Version]
  62. Gardner, A.J.; Howell, D.R.; Iverson, G.L. National Rugby League Match Scheduling and Rate of Concussion. J. Sci. Med. Sport 2019, 22, 780–783. [Google Scholar] [CrossRef] [PubMed]
  63. Savage, J.; Hooke, C.; Orchard, J.; Parkinson, R. The Incidence of Concussion in a Professional Australian Rugby League Team, 1998–2012. J. Sports Med. 2013, 2013, 304576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cosgrave, M.; Williams, S. The Epidemiology of Concussion in Professional Rugby Union in Ireland. Phys. Ther. Sport 2019, 35, 99–105. [Google Scholar] [CrossRef] [PubMed]
  65. Hinton-Bayre, A.D.; Geffen, G.; Friis, P. Presentation and Mechanisms of Concussion in Professional Rugby League Football. J. Sci. Med. Sport 2004, 7, 400–404. [Google Scholar] [CrossRef]
  66. Gabbett, T.J. Influence of Training and Match Intensity on Injuries in Rugby League. J. Sports Sci. 2004, 22, 409–417. [Google Scholar] [CrossRef]
  67. Kemp, S.; West, S.; Brooks, J.; Cross, M.; Williams, S.; Anstiss, T.; Smith, A.; Bryan, R.; Hibbins-Butler, R.; O’Leary, B.; et al. The Professional Rugby Injury Surveillance Project (PRISP) Annual Report(s) 2016/17; Rugby Football Union: Twickenham, UK, 2018. [Google Scholar]
  68. Bigler, E. Anterior and Middle Cranial Fossa in Traumatic Brain Injury: Relevant Neuroanatomy and Neuropathology in the Study of Neuropsychological Outcome. Neuropsychology 2007, 21, 515–531. [Google Scholar] [CrossRef] [Green Version]
  69. Meaney, D.F.; Smith, D.H. Biomechanics of Concussion. Clin. Sports Med. 2011, 30, 19–31. [Google Scholar] [CrossRef] [Green Version]
  70. Post, A.; Hoshizaki, B.T. Rotational Acceleration, Brain Tissue Strain, and the Relationship to Concussion. J. Biomech. Eng. 2015, 137. [Google Scholar] [CrossRef]
  71. Hoshizaki, B.; Post, A.; Kendall, M.; Karton, C.; Brien, S. The Relationship between Head Impact Characteristics and Brain Trauma. J. Neurol. Neurophysiol. 2013, 5, 1–8. [Google Scholar] [CrossRef]
  72. McAllister, T.W. Neurobiological Consequences of Traumatic Brain Injury. Dialogues Clin. Neurosci. 2011, 13, 287–300. [Google Scholar]
  73. Meythaler, J.M.; Peduzzi, J.D.; Eleftheriou, E.; Novack, T.A. Current Concepts: Diffuse Axonal Injury–Associated Traumatic Brain Injury. Arch. Phys. Med. Rehabil. 2001, 82, 1461–1471. [Google Scholar] [CrossRef]
  74. Wu, L.C.; Nangia, V.; Bui, K.; Hammoor, B.; Kurt, M.; Hernandez, F.; Kuo, C.; Camarillo, D.B. In Vivo Evaluation of Wearable Head Impact Sensors. Ann. Biomed. Eng. 2016, 44, 1234. [Google Scholar] [CrossRef] [Green Version]
  75. Guskiewicz, K.M.; Mihalik, J.P.; Shankar, V.; Marshall, S.W.; Crowell, D.H.; Oliaro, S.M.; Ciocca, M.F.; Hooker, D.N. Measurement of head impacts in collegiate football playerS. Neurosurgery 2007, 61, 1244–1253. [Google Scholar] [CrossRef]
  76. Giza, C.C.; Hovda, D.A. The New Neurometabolic Cascade of Concussion. Neurosurgery 2014, 75, S24–S33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Giza, C.C.; Hovda, D.A. The Neurometabolic Cascade of Concussion. J. Athl. Train. 2001, 36, 228–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Bazarian, J.; 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. J. Neurotrauma 2007, 24, 1447–1459. [Google Scholar] [CrossRef] [PubMed]
  79. Blumbergs, P.; Scott, G.; Manavis, J.; Wainwright, H.; Simpson, D.; McLean, A. Staining of Amyloid Precursor Protein to Study Axonal Damage in Mild Head Injury. Lancet 1994, 344, 1055–1056. [Google Scholar] [CrossRef]
  80. Blumbergs, P.; Scott, G.; Vis, J.; Wainwright, H.; Simpson, D.; McLean, A.J. Topography of Axonal Injury as Defined by Amyloid Precursor Protein and the Sector Scoring Method in Mild and Severe Closed Head Injury. J. Neurotrauma 1995, 12, 565–572. [Google Scholar] [CrossRef]
  81. Povlishock, J.T.; Pettus, E.H. Traumatically Induced Axonal Damage: Evidence for Enduring Changes in Axolemmal Permeability with Associated Cytoskeletal Change. Acta Neurochir. Suppl. 1996, 1996, 81–86. [Google Scholar] [CrossRef]
  82. Katayama, Y.; Becker, D.P.; Tamura, T.; Hovda, D.A. Massive Increases in Extracellular Potassium and the Indiscriminate Release of Glutamate Following Concussive Brain Injury. J. Neurosurg. 1990, 73, 889–900. [Google Scholar] [CrossRef]
  83. Cantu, R.; Cantu, R. Neurologic Athletic Head and Spine Injuries; W.B. Saunders Co.: Philadelphia, PA, USA, 2000. [Google Scholar]
  84. Büki, A.; Povlishock, J. All Roads Lead to Disconnection?—Traumatic Axonal Injury Revisited. Acta Neurochir. 2006, 148, 181–194. [Google Scholar] [CrossRef] [PubMed]
  85. D’Ambrosio, R.; Maris, D.O.; Grady, M.S.; Winn, H.R.; Janigro, D. Impaired K+ Homeostasis and Altered Electrophysiological Properties of Post-Traumatic Hippocampal Gila. J. Neurosci. 1999, 19, 8152–8162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Hartings, J.A.; Strong, A.J.; Fabricius, M.; Manning, A.; Bhatia, R.; Dreier, J.P.; Mazzeo, A.T.; Tortella, F.C.; Bullock, M.R. Spreading Depolarizations and Late Secondary Insults after Traumatic Brain Injury. J. Neurotrauma 2009, 26, 1857–1866. [Google Scholar] [CrossRef] [PubMed]
  87. Gaetz, M. The Neurophysiology of Brain Injury. Clin. Neurophysiol. 2004, 115, 4–18. [Google Scholar] [CrossRef] [Green Version]
  88. Choi, D.W. Ionic Dependence of Glutamate Neurotoxicity. J. Neurosci. 1987, 7, 369–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Wang, Y.; Nelson, L.D.; Laroche, A.A.; Pfaller, A.Y.; Nencka, A.S.; Koch, K.M.; McCrea, M.A. Cerebral Blood Flow Alterations in Acute Sport-Related Concussion. J. Neurotrauma 2016, 33, 1227–1236. [Google Scholar] [CrossRef] [Green Version]
  90. Weber, J.T. Altered Calcium Signaling Following Traumatic Brain Injury. Front. Pharmacol. 2012, 3, 60. [Google Scholar] [CrossRef] [Green Version]
  91. Xiong, Y.; Peterson, P.L.; Verweij, B.H.; Vinas, F.C.; Muizelaar, J.P.; LEE, C.P. Mitochondrial Dysfunction After Experimental Traumatic Brain Injury: Combined Efficacy of SNX-111 and U-101033E. J. Neurotrauma 1998, 15, 531–544. [Google Scholar] [CrossRef]
  92. Patterson, Z.R.; Holahan, M.R. Understanding the Neuroinflammatory Response Following Concussion to Develop Treatment Strategies. Front. Cell. Neurosci. 2012, 6. [Google Scholar] [CrossRef] [Green Version]
  93. Zaitlen, N.; Kraft, P. Heritability in the Genome-Wide Association Era. Hum. Genet. 2012, 1655–1664. [Google Scholar] [CrossRef] [Green Version]
  94. Zuk, O.; Hechter, E.; Sunyaev, S.R.; Lander, E.S. The Mystery of Missing Heritability: Genetic Interactions Create Phantom Heritability. Proc. Natl. Acad. Sci. USA 2012, 109, 1193–1198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Tabor, H.K.; Risch, N.J.; Myers, R.M. Candidate-Gene Approaches for Studying Complex Genetic Traits: Practical Considerations. Nat. Rev. Genet. 2002, 3, 391–397. [Google Scholar] [CrossRef] [PubMed]
  96. Rankinen, T.; Bray, M.S.; Hagberg, J.M.; Pérusse, L.; Roth, S.M.; Wolfarth, B.; Bouchard, C. The Human Gene Map for Performance and Health-Related Fitness Phenotypes: The 2005 Update. Med. Sci. Sports Exerc. 2006, 38, 1863–1888. [Google Scholar] [CrossRef] [PubMed]
  97. Bellosta, S.; Nathan, B.; Orth, M.; Dong, L.; Mahley, R.; Pitas, R. Stable Expression and Secretion of Apolipoproteins E3 and E4 in Mouse Neuroblastoma Cells Produces Differential Effects on Neurite Outgrowth. J. Biol. Chem. 1995, 270, 27063–27071. [Google Scholar] [CrossRef] [Green Version]
  98. Holtzman, D.M.; Pitas, R.E.; Kilbridge, J.; Nathan, B.; Mahley, R.W.; Bu, G.; Schwartz, A.L. Low Density Lipoprotein Receptor-Related Protein Mediates Apolipoprotein E-Dependent Neurite Outgrowth in a Central Nervous System-Derived Neuronal Cell Line. Proc. Natl. Acad. Sci. USA 1995, 92, 9480–9484. [Google Scholar] [CrossRef] [Green Version]
  99. Namboori, P.K.K.; Vineeth, K.V.; Rohith, V.; Hassan, I.; Sekhar, L.; Sekhar, A.; Nidheesh, M. The ApoE Gene of Alzheimer’s Disease (AD). Funct. Integr. Genom. 2011, 519–522. [Google Scholar] [CrossRef]
  100. Kristman, V.L.; Tator, C.H.; Kreiger, N.; Richards, D.; Mainwaring, L.; Jaglal, S.; Tomlinson, G.; Comper, P. Does the Apolipoprotein Ε4 Allele Predispose Varsity Athletes to Concussion? A Prospective Cohort Study. Clin. J. Sport Med. 2008, 18, 322–328. [Google Scholar] [CrossRef]
  101. Lambert, J.-C.; Araria-Goumidi, L.; Myllykangas, L.; Ellis, C.; Wang, J.C.; Bullido, M.J.; Harris, J.M.; Artiga, M.J.; Hernandez, D.; Kwon, J.M.; et al. Contribution of APOE Promoter Polymorphisms to Alzheimer’s Disease Risk. Neurology 2002, 59, 59–66. [Google Scholar] [CrossRef]
  102. Lendon, C.L.; Harris, J.M.; Pritchard, A.L.; Nicoll, J.A.R.; Teasdale, G.M.; Murray, G. Genetic Variation of the APOE Promoter and Outcome after Head Injury. Neurology 2003, 61, 683–685. [Google Scholar] [CrossRef]
  103. Gabbita, S.P.; Scheff, S.W.; Menard, R.M.; Roberts, K.; Fugaccia, I.; Zemlan, F.P. Cleaved-Tau: A Biomarker of Neuronal Damage after Traumatic Brain Injury. J. Neurotrauma 2005, 22, 83–94. [Google Scholar] [CrossRef]
  104. Weingarten, M.D.; Lockwood, A.H.; Hwo, S.Y.; Kirschner, M.W. A Protein Factor Essential for Microtubule Assembly. Proc. Natl. Acad. Sci. USA 1975, 72, 1858–1862. [Google Scholar] [CrossRef] [Green Version]
  105. Wagner, O.I.; Rammensee, S.; Korde, N.; Wen, Q.; Leterrier, J.F.; Janmey, P.A. Softness, Strength and Self-Repair in Intermediate Filament Networks. Exp. Cell Res. 2007, 313, 2228–2235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Iwata, N.; Tsubuki, S.; Takaki, Y.; Watanabe, K.; Sekiguchi, M.; Hosoki, E.; Kawashima-Morishima, M.; Lee, H.J.; Hama, E.; Sekine-Aizawa, Y.; et al. Identification of the Major Aβ1-42-Degrading Catabolic Pathway in Brain Parenchyma: Suppression Leads to Biochemical and Pathological Deposition. Nat. Med. 2000, 6, 143–150. [Google Scholar] [CrossRef] [PubMed]
  107. Iwata, N.; Tsubuki, S.; Takaki, Y.; Shirotani, K.; Lu, B.; Gerard, N.P.; Gerard, C.; Hama, E.; Lee, H.J.; Saido, T.C. Metabolic Regulation of Brain Aβ by Neprilysin. Science 2001, 292, 1550–1552. [Google Scholar] [CrossRef]
  108. Johnson, V.E.; Stewart, W.; Graham, D.I.; Stewart, J.E.; Praestgaard, A.H.; Smith, D.H. A Neprilysin Polymorphism and Amyloid-β Plaques after Traumatic Brain Injury. J. Neurotrauma 2009, 26, 1197–1202. [Google Scholar] [CrossRef] [Green Version]
  109. Lipsky, R.H.; Marini, A.M. Brain-Derived Neurotrophic Factor in Neuronal Survival and Behavior-Related Plasticity. Ann. N. Y. Acad. Sci. 2007, 1122, 130–143. [Google Scholar] [CrossRef] [PubMed]
  110. McAllister, A.K.; Lo, D.C.; Katz, L.C. Neurotrophins Regulate Dendritic Growth in Developing Visual Cortex. Neuron 1995, 15, 791–803. [Google Scholar] [CrossRef] [Green Version]
  111. Lu, B. Pro-Region of Neurotrophins: Role in Synaptic Modulation. Neuron 2003, 39, 735–738. [Google Scholar] [CrossRef] [Green Version]
  112. Ali, F.; Meier, R. Primate Home Range and GRIN2A, a Receptor Gene Involved in Neuronal Plasticity: Implications for the Evolution of Spatial Memory. Genes Brain Behav. 2009, 8, 435–441. [Google Scholar] [CrossRef]
  113. De Quervain, D.J.F.; Papassotiropoulos, A. Identification of a Genetic Cluster Influencing Memory Performance and Hippocampal Activity in Humans. Proc. Natl. Acad. Sci. USA 2006, 103, 4270–4274. [Google Scholar] [CrossRef] [Green Version]
  114. Itokawa, M.; Yamada, K.; Iwayama-Shigeno, Y.; Ishitsuka, Y.; Detera-Wadleigh, S.; Yoshikawa, T. Genetic Analysis of a Functional GRIN2A Promoter (GT)n Repeat in Bipolar Disorder Pedigrees in Humans. Neurosci. Lett. 2003, 345, 53–56. [Google Scholar] [CrossRef]
  115. Itokawa, M.; Yamada, K.; Yoshitsugu, K.; Toyota, T.; Suga, T.; Ohba, H.; Watanabe, A.; Hattori, E.; Shimizu, H.; Kumakura, T.; et al. A Microsatellite Repeat in the Promoter of the N-Methyl-D-Aspartate Receptor 2A Subunit (GRIN2A) Gene Suppresses Transcriptional Activity and Correlates with Chronic Outcome in Schizophrenia. Pharmacogenetics 2003, 13, 271–278. [Google Scholar] [CrossRef] [PubMed]
  116. Lipsky, R.H.; Sparling, M.B.; Ryan, L.M.; Xu, K.; Salazar, A.M.; Goldman, D.; Warden, D.L. Association of COMT Val158Met Genotype with Executive Functioning Following Traumatic Brain Injury. J. Neuropsychiatry Clin. Neurosci. 2005, 17, 465–471. [Google Scholar] [CrossRef] [PubMed]
  117. Gallinat, J.; Bajbouj, M.; Sander, T.; Schlattmann, P.; Xu, K.; Ferro, E.F.; Goldman, D.; Winterer, G. Association of the G1947A COMT (Val(108/158)Met) Gene Polymorphism with Prefrontal P300 during Information Processing. Biol. Psychiatry 2003, 54, 40–48. [Google Scholar] [CrossRef]
  118. Chen, J.; Lipska, B.; Halim, N.; Ma, Q.; Matsumoto, M.; Melhem, S.; Kolachana, B.; Hyde, T.; Herman, M.; Apud, J.; et al. Functional Analysis of Genetic Variation in Catechol-O-Methyltransferase (COMT): Effects on MRNA, Protein, and Enzyme Activity in Postmortem Human Brain. Am. J. Hum. Genet. 2004, 75, 807–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Nymberg, C.; Banaschewski, T.; Bokde, A.L.; Büchel, C.; Conrod, P.; Flor, H.; Frouin, V.; Garavan, H.; Gowland, P.; Heinz, A.; et al. DRD2/ANKK1 Polymorphism Modulates the Effect of Ventral Striatal Activation on Working Memory Performance. Neuropsychopharmacology 2014, 39, 2357–2365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Vijayraghavan, S.; Wang, M.; Birnbaum, S.G.; Williams, G.V.; Arnsten, A.F. Inverted-U Dopamine D1 Receptor Actions on Prefrontal Neurons Engaged in Working Memory. Nat. Neurosci. 2007, 10, 376–384. [Google Scholar] [CrossRef]
  121. Neville, M.J.; Johnstone, E.C.; Walton, R.T. Identification and Characterization of ANKK1: A Novel Kinase Gene Closely Linked to DRD2 on Chromosome Band 11q23.1. Hum. Mutat. 2004, 23, 540–545. [Google Scholar] [CrossRef]
  122. Jönsson, E.G.; Nöthen, M.M.; Grünhage, F.; Farde, L.; Nakashima, Y.; Propping, P.; Sedvall, G.C. Polymorphisms in the Dopamine D2 Receptor Gene and Their Relationships to Striatal Dopamine Receptor Density of Healthy Volunteers. Mol. Psychiatry 1999, 4, 290–296. [Google Scholar] [CrossRef] [Green Version]
  123. Ritchie, T.; Noble, E.P. Association of Seven Polymorphisms of the D2 Dopamine Receptor Gene with Brain Receptor-Binding Characteristics. Neurochem. Res. 2003, 28, 73–82. [Google Scholar] [CrossRef]
  124. Thompson, J.; Thomas, N.; Singleton, A.; Piggott, M.; Lloyd, S.; Perry, E.K.; Morris, C.M.; Perry, R.H.; Ferrier, I.N.; Court, J.A. D2 Dopamine Receptor Gene (DRD2) Taq 1 A Polymorphism: Reduced Dopamine D2 Receptor Binding in the Human Striatum Associated with the A1 Allele. Pharmacogenetics 1997, 7, 479–484. [Google Scholar] [CrossRef] [PubMed]
  125. Okuyama, Y.; Ishiguro, H.; Nankai, M.; Shibuya, H.; Watanabe, A.; Arinami, T. Identification of a Polymorphism in the Promoter Region of DRD4 Associated with the Human Novelty Seeking Personality Trait. Mol. Psychiatry 2000, 5, 64–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hamidovic, A.; Dlugos, A.; Skol, A.; Palmer, A.A.; de Wit, H. Evaluation of Genetic Variability in the Dopamine Receptor D2 in Relation to Behavioral Inhibition and Impulsivity/Sensation Seeking: An Exploratory Study with d-Amphetamine in Healthy Participants. Exp. Clin. Psychopharmacol. 2009, 17, 374–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Balestri, M.; Calati, R.; Serretti, A.; De Ronchi, D. Genetic Modulation of Personality Traits. Int. Clin. Psychopharmacol. 2014, 29, 1–15. [Google Scholar] [CrossRef]
  128. Heils, A.; Teufel, A.; Petri, S.; Stöber, G.; Riederer, P.; Bengel, D.; Lesch, K.P. Allelic Variation of Human Serotonin Transporter Gene Expression. J. Neurochem. 2002, 66, 2621–2624. [Google Scholar] [CrossRef]
  129. Lesch, K.P.; Bengel, D.; Heils, A.; Sabol, S.Z.; Greenberg, B.D.; Petri, S.; Benjamin, J.; Müller, C.R.; Hamer, D.H.; Murphy, D.L. Association of Anxiety-Related Traits with a Polymorphism in the Serotonin Transporter Gene Regulatory Region. Science 1996, 274, 1527–1531. [Google Scholar] [CrossRef]
  130. Stoltenberg, S.F.; Lehmann, M.K.; Anderson, C.; Nag, P.; Anagnopoulos, C. Serotonin Transporter (5-HTTLPR) Genotype and Childhood Trauma Are Associated with Individual Differences in Decision Making. Front. Genet. 2011, 2. [Google Scholar] [CrossRef] [Green Version]
  131. Sonuga-Barke, E.J.S.; Kumsta, R.; Schlotz, W.; Lasky-Su, J.; Marco, R.; Miranda, A.; Mulas, F.; Oades, R.D.; Banaschewski, T.; Mueller, U.; et al. A Functional Variant of the Serotonin Transporter Gene ((SLC6A4) Moderates Impulsive Choice in Attention-Deficit/Hyperactivity Disorder Boys and Siblings. Biol. Psychiatry 2011, 70, 230–236. [Google Scholar] [CrossRef] [Green Version]
  132. Garry, P.; Ezra, M.; Rowland, M.; Westbrook, J.; Pattinson, K. The Role of the Nitric Oxide Pathway in Brain Injury and Its Treatment—From Bench to Bedside. Exp. Neurol. 2015, 263, 235–243. [Google Scholar] [CrossRef] [Green Version]
  133. Toda, N.; Ayajiki, K.; Okamura, T. Cerebral Blood Flow Regulation by Nitric Oxide: Recent Advances. Pharmacol. Rev. 2009, 61, 62–97. [Google Scholar] [CrossRef] [Green Version]
  134. Ahn, M.J.; Sherwood, E.R.; Prough, D.S.; Cheng, Y.L.; DeWitt, D.S. The Effects of Traumatic Brain Injury on Cerebral Blood Flow and Brain Tissue Nitric Oxide Levels and Cytokine Expression. J. Neurotrauma 2004, 21, 1431–1442. [Google Scholar] [CrossRef] [PubMed]
  135. Tuzgen, S.; Tanriover, N.; Uzan, M.; Tureci, E.; Tanriverdi, T.; Gumustas, K.; Kuday, C. Nitric Oxide Levels in Rat Cortex, Hippocampus, Cerebellum, and Brainstem after Impact Acceleration Head Injury. Neurol. Res. 2003, 25, 31–34. [Google Scholar] [CrossRef] [PubMed]
  136. Asif, A.R.; Oellerich, M.; Armstrong, V.W.; Hecker, M.; Cattaruzza, M. T-786C Polymorphism of the Nos-3 Gene and the Endothelial Cell Response to Fluid Shear Stress—A Proteome Analysis. J. Proteome Res. 2009, 8, 3161–3168. [Google Scholar] [CrossRef] [PubMed]
  137. Robertson, C.S.; Gopinath, S.P.; Valadka, A.B.; Van, M.; Swank, P.R.; Goodman, J.C. Variants of the Endothelial Nitric Oxide Gene and Cerebral Blood Flow after Severe Traumatic Brain Injury. J. Neurotrauma 2011, 28, 727–737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Sayed-Tabatabaei, F.A.; Oostra, B.A.; Isaacs, A.; Van Duijn, C.M.; Witteman, J.C.M. ACE Polymorphisms. Circ. Res. 2006, 1123–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ariza, M.; del Matarin, M.; Junqué, C.; Mataró, M.; Clemente, I.; Moral, P.; Antonia Poca, M.; Garnacho, A.; Sahuquillo, J. Influence of Angiotensin-Converting Enzyme Polymorphism on Neuropsychological Subacute Performance in Moderate and Severe Traumatic. J. Neuropsychiatry Clin. Neurosci. 2006, 18, 39–44. [Google Scholar] [CrossRef] [PubMed]
  140. Dardiotis, E.; Paterakis, K.; Siokas, V.; Tsivgoulis, G.; Dardioti, M.; Grigoriadis, S.; Simeonidou, C.; Komnos, A.; Kapsalaki, E.; Fountas, K.; et al. Effect of Angiotensin-Converting Enzyme Tag Single Nucleotide Polymorphisms on the Outcome of Patients with Traumatic Brain Injury. Pharm. Genom. 2015, 25, 485–490. [Google Scholar] [CrossRef]
  141. Oshima, T.; Lee, S.; Sato, A.; Oda, S.; Hirasawa, H.; Yamashita, T. TNF-α Contributes to Axonal Sprouting and Functional Recovery Following Traumatic Brain Injury. Brain Res. 2009, 1290, 102–110. [Google Scholar] [CrossRef]
  142. Bermpohl, D.; You, Z.; Korsmeyer, S.J.; Moskowitz, M.A.; Whalen, M.J. Traumatic Brain Injury in Mice Deficient in Bid: Effects on Histopathology and Functional Outcome. J. Cereb. Blood Flow Metab. 2006, 26, 625–633. [Google Scholar] [CrossRef] [Green Version]
  143. Waters, R.J.; Murray, G.D.; Teasdale, G.M.; Stewart, J.; Day, I.; Lee, R.J.; Nicoll, J.A.R. Cytokine Gene Polymorphisms and Outcome after Traumatic Brain Injury. J. Neurotrauma 2013, 30, 1710–1716. [Google Scholar] [CrossRef] [Green Version]
  144. Letterio, J.J.; Roberts, A.B. Regulation of immune responses by TGF-β. Annu. Rev. Immunol. 1998, 16, 137–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Shah, R.; Hurley, C.K.; Posch, P.E. A Molecular Mechanism for the Differential Regulation of TGF-Β1 Expression Due to the Common SNP −509C-T (c. -1347C > T). Hum. Genet. 2006, 120, 461–469. [Google Scholar] [CrossRef] [PubMed]
  146. Uzan, M.; Tanriverdi, T.; Baykara, O.; Kafadar, A.; Sanus, G.Z.; Tureci, E.; Ozkara, C.; Uysal, O.; Buyra, N. Association between Interleukin-1 Beta (IL-1β) Gene Polymorphism and Outcome after Head Injury: An Early Report. Acta Neurochir. 2005, 147, 715–720. [Google Scholar] [CrossRef] [PubMed]
  147. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The Pro- and Anti-Inflammatory Properties of the Cytokine Interleukin-6. Biochim. Biophys. Acta 2011, 1813, 878–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Weisgraber, K.H. Apolipoprotein E: Structure-Function Relationships. Adv. Protein Chem. 1994, 45, 249–302. [Google Scholar] [CrossRef] [PubMed]
  149. Abrahams, S.; Mc Fie, S.; Patricios, J.; Sutere, J.; Posthumus, M.; Septembera, A.V. An Association between Polymorphisms within the APOE Gene and Concussion Aetiology in Rugby Union Players. J. Sci. Med. Sport 2018, 21, 117–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Jordan, B.D.; Relkin, N.R.; Ravdin, L.D.; Jacobs, A.R.; Bennett, A.; Gandy, S. Apolipoprotein E Ε4 Associated with Chronic Traumatic Brain Injury in Boxing. J. Am. Med. Assoc. 1997, 278, 136–140. [Google Scholar] [CrossRef]
  151. Lawrence, D.W.; Comper, P.; Hutchison, M.G.; Sharma, B. The Role of Apolipoprotein E Episilon (ε)-4 Allele on Outcome Following Traumatic Brain Injury: A Systematic Review. Brain Inj. 2015, 29, 1018–1031. [Google Scholar] [CrossRef]
  152. Merritt, V.; Arnett, P. Apolipoprotein E (APOE) Ε4 Allele Is Associated with Increased Symptom Reporting Following Sports Concussion. J. Int. Neuropsychol. Soc. 2016, 22, 89–94. [Google Scholar] [CrossRef]
  153. Bogoslovsky, T.; Wilson, D.; Chen, Y.; Hanlon, D.; Gill, J.; Jeromin, A.; Song, L.; Moore, C.; Gong, Y.; Kenney, K.; et al. Increases of Plasma Levels of Glial Fibrillary Acidic Protein, Tau, and Amyloid β up to 90 Days after Traumatic Brain Injury. J. Neurotrauma 2017, 34, 66–73. [Google Scholar] [CrossRef] [Green Version]
  154. Turner, R.C.; Lucke-Wold, B.P.; Robson, M.J.; Omalu, B.I.; Petraglia, A.L.; Bailes, J.E. Repetitive Traumatic Brain Injury and Development of Chronic Traumatic Encephalopathy: A Potential Role for Biomarkers in Diagnosis, Prognosis, and Treatment? Front. Neurol. 2012, 3, 186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. De Silva, R.; Lashley, T.; Strand, C.; Shiarli, A.-M.; Shi, J.; Tian, J.; Bailey, K.L.; Davies, P.; Bigio, E.H.; Arima, K.; et al. An Immunohistochemical Study of Cases of Sporadic and Inherited Frontotemporal Lobar Degeneration Using 3R- and 4R-Specific Tau Monoclonal Antibodies. Acta Neuropathol. 2006, 111, 329–340. [Google Scholar] [CrossRef] [PubMed]
  156. D’Souza, I.; Schellenberg, G.D. Regulation of Tau Isoform Expression and Dementia. Biochim. Biophys. Acta 2005, 1739, 104–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Togo, T.; Sahara, N.; Yen, S.-H.; Cookson, N.; Ishizawa, T.; Hutton, M.; de Silva, R.; Lees, A.; Dickson, D.W. Argyrophilic Grain Disease Is a Sporadic 4-Repeat Tauopathy. J. Neuropathol. Exp. Neurol. 2002, 61, 547–556. [Google Scholar] [CrossRef] [Green Version]
  158. Sergeant, N.; Wattez, A.; Delacourte, A. Neurofibrillary Degeneration in Progressive Supranuclear Palsy and Corticobasal Degeneration: Tau Pathologies with Exclusively ‘Exon 10’ Isoforms. J. Neurochem. 1999, 72, 1243–1249. [Google Scholar] [CrossRef] [Green Version]
  159. Couchie, D.; Mavilia, C.; Georgieff, I.S.; Liem, R.K.; Shelanski, M.L.; Nunez, J. Primary Structure of High Molecular Weight Tau Present in the Peripheral Nervous System. Proc. Natl. Acad. Sci. USA 1992, 89, 4378–4381. [Google Scholar] [CrossRef] [Green Version]
  160. Poorkaj, P.; Bird, T.D.; Wijsman, E.; Nemens, E.; Garruto, R.M.; Anderson, L.; Andreadis, A.; Wiederholt, W.C.; Raskind, M.; Schellenberg, G.D. Tau Is a Candidate Gene for Chromosome 17 Frontotemporal Dementia. Ann. Neurol. 1998, 43, 815–825. [Google Scholar] [CrossRef]
  161. Mcdevitt, J.K.; Tierney, R.T.; Mansell, J.L.; Driban, J.B.; Higgins, M.; Toone, N.; Mishra, A.; Krynetskiy, E. Brain Injury Neuronal Structural Protein Polymorphism and Concussion in College Athletes Neuronal Structural Protein Polymorphism and Concussion in College Athletes. Brain Inj. 2011, 25, 1108–1113. [Google Scholar] [CrossRef]
  162. Egan, M.F.; Kojima, M.; Callicott, J.H.; Goldberg, T.E.; Kolachana, B.S.; Bertolino, A.; Zaitsev, E.; Gold, B.; Goldman, D.; Dean, M.; et al. The BDNF Val66met Polymorphism Affects Activity-Dependent Secretion of BDNF and Human Memory and Hippocampal Function. Cell 2003, 112, 257–269. [Google Scholar] [CrossRef] [Green Version]
  163. Felderhoff-Mueser, U.; Sifringer, M.; Pesditschek, S.; Kuckuck, H.; Moysich, A.; Bittigau, P.; Ikonomidou, C. Pathways Leading to Apoptotic Neurodegeneration Following Trauma to the Developing Rat Brain. Neurobiol. Dis. 2002, 11, 231–245. [Google Scholar] [CrossRef] [Green Version]
  164. Oyesiku, N.M.; Evans, C.O.; Houston, S.; Darrell, R.S.; Smith, J.S.; Fulop, Z.L.; Dixon, C.E.; Stein, D.G. Regional Changes in the Expression of Neurotrophic Factors and Their Receptors Following Acute Traumatic Brain Injury in the Adult Rat Brain. Brain Res. 1999, 833, 161–172. [Google Scholar] [CrossRef]
  165. Hicks, R.R.; Numan, S.; Dhillon, H.S.; Prasad, M.R.; Seroogy, K.B. Alterations in BDNF and NT-3 MRNAs in Rat Hippocampus after Experimental Brain Trauma. Mol. Brain Res. 1997, 48, 401–406. [Google Scholar] [CrossRef]
  166. Dretsch, M.N.; Williams, K.; Emmerich, T.; Crynen, G.; Ait-Ghezala, G.; Chaytow, H.; Mathura, V.; Crawford, F.C.; Iverson, G.L. Brain-Derived Neurotropic Factor Polymorphisms, Traumatic Stress, Mild Traumatic Brain Injury, and Combat Exposure Contribute to Postdeployment Traumatic Stress. Brain Behav. 2016, 6, e00392. [Google Scholar] [CrossRef] [Green Version]
  167. Narayanan, V.; Veeramuthu, V.; Ahmad-Annuar, A.; Ramli, N.; Waran, V.; Chinna, K.; Bondi, M.W.; Delano-Wood, L.; Ganesan, D. Missense Mutation of Brain Derived Neurotrophic Factor (BDNF) Alters Neurocognitive Performance in Patients with Mild Traumatic Brain Injury: A Longitudinal Study. PLoS ONE 2016, 11, e0158838. [Google Scholar] [CrossRef] [PubMed]
  168. McDevitt, J.; Tierney, R.T.; Phillips, J.; Gaughan, J.P.; Torg, J.S.; Krynetskiy, E. Association between GRIN2A Promoter Polymorphism and Recovery from Concussion. Brain Inj. 2015, 29, 1674–1681. [Google Scholar] [CrossRef]
  169. Willmott, C.; Withiel, T.; Ponsford, J.; Burke, R. COMT Val158Met and Cognitive and Functional Outcomes after Traumatic Brain Injury. J. Neurotrauma 2014, 31, 1507–1514. [Google Scholar] [CrossRef]
  170. Riba, J.; Krämer, U.; Heldmann, M.; Richter, S.; Münte, T. Dopamine Agonist Increases Risk Taking but Blunts Reward-Related Brain Activity. PLoS ONE 2008, 3, e2479. [Google Scholar] [CrossRef] [Green Version]
  171. Dalley, J.; Roiser, J. Dopamine, Serotonin and Impulsivity. Neuroscience 2012, 215, 42–58. [Google Scholar] [CrossRef] [Green Version]
  172. McAllister, T.W.; Rhodes, C.H.; Flashman, L.A.; McDonald, B.C.; Belloni, D.; Saykin, A.J. Effect of the Dopamine D2 Receptor T Allele on Response Latency after Mild Traumatic Brain Injury. Am. J. Psychiatry 2005, 162, 1749–1751. [Google Scholar] [CrossRef]
  173. McAllister, T.W.; Flashman, L.A.; Harker Rhodes, C.; Tyler, A.L.; Moore, J.H.; Saykin, A.J.; McDonald, B.C.; Tosteson, T.D.; Tsongalis, G.J. Single Nucleotide Polymorphisms in ANKK1 and the Dopamine D2 Receptor Gene Affect Cognitive Outcome Shortly after Traumatic Brain Injury: A Replication and Extension Study. Brain Inj. 2008, 22, 705–714. [Google Scholar] [CrossRef] [Green Version]
  174. Yue, J.K.; Pronger, A.M.; Ferguson, A.R.; Temkin, N.R.; Sharma, S.; Rosand, J.; Sorani, M.D.; McAllister, T.W.; Barber, J.; Winkler, E.A.; et al. Association of a Common Genetic Variant within ANKK1 with Six-Month Cognitive Performance after Traumatic Brain Injury. Neurogenetics 2015, 16, 169–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Zhang, Y.; Bertolino, A.; Fazio, L.; Blasi, G.; Rampino, A.; Romano, R.; Lee, M.T.; Xiao, T.; Papp, A.; Wang, D.; et al. Polymorphisms in Human Dopamine D2 Receptor Gene Affect Gene Expression, Splicing, and Neuronal Activity during Working Memory. Source 2007, 104, 20552–20557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Abrahams, S.; Mcfie, S.; Lacerda, M.; Patricios, J.; Suter, J.; September, A.V.; Posthumus, M. Unravelling the Interaction between the DRD2 and DRD4 Genes, Personality Traits and Concussion Risk. BMJ Open Sport Exerc. Med. 2019, 5, 465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Hu, X.Z.; Lipsky, R.H.; Zhu, G.; Akhtar, L.A.; Taubman, J.; Greenberg, B.D.; Xu, K.; Arnold, P.D.; Richter, M.A.; Kennedy, J.L.; et al. Serotonin Transporter Promoter Gain-of-Function Genotypes Are Linked to Obsessive-Compulsive Disorder. Am. J. Hum. Genet. 2006, 78, 815–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Zubkov, A.Y.; Pilkington, A.S.; Bernanke, D.H.; Parent, A.D.; Zhang, J. Posttraumatic Cerebral Vasospasm: Clinical and Morphological Presentations. J. Neurotrauma 1999, 16, 763–770. [Google Scholar] [CrossRef] [PubMed]
  179. Chodobski, A.; Zink, B.J.; Szmydynger-Chodobska, J. Blood-Brain Barrier Pathophysiology in Traumatic Brain Injury. Transl Stroke Res. 2011, 2, 492–516. [Google Scholar] [CrossRef] [Green Version]
  180. Shohami, E.; Beit-Yannai, E.; Horowitz, M.; Kohen, R. Oxidative Stress in Closed-Head Injury: Brain Antioxidant Capacity as an Indicator of Functional Outcome. J. Cereb. Blood Flow Metab. 1997, 1007–1019. [Google Scholar] [CrossRef] [Green Version]
  181. Kroeger, K.M.; Carville, K.S.; Abraham, L.J. The -308 Tumor Necrosis Factor-α Promoter Polymorphism Effects Transcription. Mol. Immunol. 1997, 34, 391–399. [Google Scholar] [CrossRef]
  182. Dalgard, C.L.; Cole, J.T.; Kean, W.S.; Lucky, J.J.; Sukumar, G.; McMullen, D.C.; Pollard, H.B.; Watson, W.D. The Cytokine Temporal Profile in Rat Cortex after Controlled Cortical Impact. Front. Mol. Neurosci. 2012, 5. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Sequence of events and possible recovery durations post-concussion.
Figure 1. Sequence of events and possible recovery durations post-concussion.
Sports 09 00019 g001
Figure 2. Concussive event leading to the neurometabolic cascade. Glut, glutamate; K+, potassium; Ca2+, calcium; Mg2+, magnesium; AMPA, α -amino-3-hydroxy-5methyl-4-isoxazole-propionic acid.
Figure 2. Concussive event leading to the neurometabolic cascade. Glut, glutamate; K+, potassium; Ca2+, calcium; Mg2+, magnesium; AMPA, α -amino-3-hydroxy-5methyl-4-isoxazole-propionic acid.
Sports 09 00019 g002
Table 1. Candidate genes linked to TBI.
Table 1. Candidate genes linked to TBI.
Gene NameGene AbbreviationPolymorphism IdentifierRelevant Effects Associated with TBI
Apolipoprotein EAPOErs429358
rs7412
rs405509
Affects repair and plasticity of the brain [97,98]. APOE isoforms have differing effects on neurite extension, which can influence ability to recover post-concussion [97,98,99,100].Associated with functional regulation of APOE transcription [101,102].
Microtubule-associated protein tauMAPTrs10445337
rs2435211
rs2435200
Affects repair and plasticity of the brain via modulation of microtubule formation, structural stabilisation of the neuronal axons and drives growth of neurites [103,104].
Neurofilament heavyNEFHrs165602Affects repair and plasticity of the brain via modulation of the neuronal cytoskeleton is to resist the resultant strain caused by biomechanical forces [105].
Membrane metalloendopeptidaseMMEGT repeat promoter polymorphism of neprilysinAffects repair and plasticity of the brain as this gene encodes for the neprilysin protease which degrades Aβ proteins [106,107,108].
Brain-derived neurotrophic factorantisense RNABDNF-ASrs6265Affects repair and plasticity of the brain via strengthening existing synaptic connections and modulating the creation of new synapses [109,110,111].
Glutamate ionotropic receptor NMDA type subunit 2A promoterGRIN2Ars3219790Affects duration of concussion via potential modulation of glutamate-gated ion channel proteins [112,113,114,115].
Catechol-O-methyltransferaseCOMTrs4680Affects cognitive behavioural capacity post-concussion and could increase impulsivity and risk taking [116,117,118].
Ankyrin repeat and kinase domain containing 1ANKK1rs1800497Affects cognitive behavioural capacity via modulation of expression of D2 receptors [119,120,121,122,123,124].
Dopamine receptor D2
Dopamine receptor D4
DRD2
DRD4
rs12364283
rs1076560
rs1800955
Affects personality traits, associated with risk-taking behaviours (impulsivity, behavioural inhibition and novelty seeking) [125,126].
Solute carrier family 6 member 4SLC6A4rs4795541
rs25531
Reported to play a role in personality and behavior via increased harm avoidance and impulsivity behaviours [127,128,129,130,131].
Endothelial nitric oxide synthaseNOS3rs2070744Could affect severity of concussion and cognitive behavioural capacity post-concussion via modulation of cerebral vasospasm [132,133,134,135,136,137].
Angiotensin I-converting enzymeACErs4646994
rs7221780
rs8066276
Affects cognitive behavioural capacity post-concussion via modulation of cerebral blood flow [138,139,140].
Tumour necrosis factorTNFrs1800629
rs1800468
rs1800469
Could affect neuroinflammation and severity of concussion [141,142,143].
Transforming growth factor beta 1TGFB1rs1800468
rs1800469
Regulation of the anti-inflammatory mediator TGFB1 could affect severity of concussion [144,145].
Interleukin 1 alpha
interleukin 1 beta
IL1A
IL1B
rs1800587
rs16944
rs1143634
Affects severity of TBI via potential modulation of the inflammatory process and secondary conditions [146].
Interleukin 6 receptorIL6Rrs2228145Affects severity of concussion potential via modulation of the inflammatory process and cognitive behavioural capacity post-concussion [147].
Table 2. Three isoforms and six possible genotypes of APOE.
Table 2. Three isoforms and six possible genotypes of APOE.
APOE IsoformAPOE Genotypers429358rs7412
ε2 TT
ε3 TC
ε4 CC
ε2/ε2TTTT
ε2/ε3TTCT
ε2/ε4CTCT
ε3/ε3TTCC
ε3/ε4CTCC
ε4/ε4CCCC
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Antrobus, M.R.; Brazier, J.; Stebbings, G.K.; Day, S.H.; Heffernan, S.M.; Kilduff, L.P.; Erskine, R.M.; Williams, A.G. Genetic Factors That Could Affect Concussion Risk in Elite Rugby. Sports 2021, 9, 19. https://0-doi-org.brum.beds.ac.uk/10.3390/sports9020019

AMA Style

Antrobus MR, Brazier J, Stebbings GK, Day SH, Heffernan SM, Kilduff LP, Erskine RM, Williams AG. Genetic Factors That Could Affect Concussion Risk in Elite Rugby. Sports. 2021; 9(2):19. https://0-doi-org.brum.beds.ac.uk/10.3390/sports9020019

Chicago/Turabian Style

Antrobus, Mark R., Jon Brazier, Georgina K. Stebbings, Stephen H. Day, Shane M. Heffernan, Liam P. Kilduff, Robert M. Erskine, and Alun G. Williams. 2021. "Genetic Factors That Could Affect Concussion Risk in Elite Rugby" Sports 9, no. 2: 19. https://0-doi-org.brum.beds.ac.uk/10.3390/sports9020019

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop