Open Access

Traumatic brain injury: Advances in coagulopathy (Review)

  • Authors:
    • Hongqiao Hou
    • Zhe Qu
    • Ruping Liu
    • Bowen Jiang
    • Lanlan Wang
    • Aiqun Li
  • View Affiliations

  • Published online on: August 27, 2024     https://doi.org/10.3892/br.2024.1844
  • Article Number: 156
  • Copyright: © Hou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Trauma is a prevalent cause of coagulopathy, with traumatic brain injury (TBI) accompanied by coagulation disorders particularly linked to adverse outcomes. TBI is distinguished by minimal bleeding volume and unique injury sites, which precipitate complex coagulation disturbances. Historically, research into trauma‑induced coagulopathy has primarily concentrated on the molecular biology and pathophysiology of endogenous anticoagulation and inflammation. Nonetheless, recognizing that cells are the fundamental units of structure and function in all living organisms, the present review aimed to distill our understanding of coagulopathy post‑TBI by elucidating the intricate cellular mechanisms involving endothelial cells, neutrophils and platelets. Additionally, this study evaluates the strengths and weaknesses of various diagnostic tools and discusses the characteristics of pharmacological treatments and potential therapies for patients with TBI and coagulation disorders. The aim of this review is to amalgamate recent updates in mechanistic research and innovative diagnostic and therapeutic methodologies, thereby fostering the progression of precision medicine within this specialized domain.

1. Introduction

Traumatic brain injury (TBI) encompasses a series of complex pathological processes wherein brain tissue structure and function are compromised due to mechanical force applied to the head, stemming from various causes. Of considerable interest is the potential link between genetic modifications and an augmented susceptibility to TBI. Specifically, the polymorphisms within the apolipoprotein E promoter region, the TAU gene (1) and the brain-derived neurotrophic factor gene have been implicated in elevating the risk of concussion and contributing to adverse outcomes subsequent to a TBI episode. Additionally, alterations in the dopamine receptor D2 gene may potentiate brain injury risk by impacting cognitive functions and behavioral traits in affected individuals. Nevertheless, the precise contribution of NEFH gene mutations to the pathology of concussion remains an area requiring further elucidation through research (2).

The incidence and mortality rates of TBI are notably higher in low- and middle-income countries compared with high-income countries (3), affecting an estimated 10 million individuals worldwide annually (4). Falls are the predominant cause of TBI, especially among individuals >50 years old (5). This demographic often presents with comorbidities and may be using anticoagulant medications prior to injury, factors that amplify the complexity and mortality risk associated with coagulation abnormalities post-TBI (6). Coagulopathy in patients with TBI typically manifests as abnormalities in conventional coagulation assays. However, the prevalence of early coagulopathy varies widely due to inconsistent definitions and differences in the severity of the injury (7).

Most patients with severe TBI demonstrate abnormal coagulation test results shortly after injury, whereas this is less common in those with mild injuries (8). The severity of TBI is generally assessed utilizing the Glasgow Coma Scale, categorizing injuries as mild (14-15 points), moderate (9-13 points) and severe (3-8 points) (9). The mortality rate for severe TBI cases is approximately one-third, and ~60% of survivors suffer from enduring physical, mental and social deficits (10). Patients with TBI accompanied by coagulopathy are often closely associated with poor prognosis (11), thus advancing research and treatment for coagulopathy following TBI is of paramount importance.

In the human body, cells are the fundamental units of structure, function and biological processes. The present review updated and simplified the series of coagulopathy-related events occurring after TBI by summarizing the mechanistic changes and roles of three types of cells [endothelial cells (ECs), neutrophils and platelets] in the coagulation process post-TBI. Additionally, by consolidating the pharmacological treatments and potential therapeutic applications for patients with TBI with coagulation dysfunction, the present study aimed for this review to provide more insights into precision medicine by summarizing updated mechanistic studies and innovative diagnostic and therapeutic techniques. To facilitate a better understanding of the content for the readers, definitions for the specialized terminology mentioned in this article have been provided (Table I).

Table I

Definitions of specialized terminology.

Table I

Definitions of specialized terminology.

Specialized terminologyExplanation
Gaussian distribution patternA pattern of injury where the brain tissue at the center of the trauma experiences the highest and most severe damage, with the severity of the injury diminishing radially outward.
Brain-derived cell microvesiclesSmall vesicles secreted by brain tissue cells and vascular endothelial cells containing biomolecules such as proteins, DNA and RNA from their cell of origin.
LactoferrinA member of the transferrin family with antioxidant and anti-inflammatory properties.
Neutrophil extracellular trapsWeb-like structures released extracellularly by activated neutrophils containing components such as DNA and histones that have the ability to trap and kill pathogens among other functions.
Damage-associated molecular patternsEndogenous molecules released by host cells upon injury which include high mobility group box 1, DNA and RNA, triggering immune and inflammatory responses.
Platelet-derived extracellular vesiclesVesicles released by platelets through budding or exocytosis, participating in processes such as inflammation and immunomodulation. Platelet-derived exosomes represent a specific subset of these extracellular vesicles.
Multimodality monitoringA comprehensive monitoring method that combines multiple monitoring technologies and parameters to assess the functional status of the brain. This approach aims to provide an integrated view of cerebral pathophysiology in real-time.

2. Cellular mechanisms of coagulopathy following TBI

The mechanisms underpinning coagulopathy subsequent to TBI are multifaceted, encompassing a broad spectrum of cellular alterations. These primarily include alterations in ECs, neutrophils and platelets (Fig. 1).

Endothelium and blood-brain barrier (BBB)

The endothelium and BBB are pivotal in maintaining hemostatic balance. Under normal conditions, the ECs function as a barrier to prevent the release of procoagulant factors, such as cerebral microvesicles, into the circulation (12,13). However, these cells can sustain rapid damage during the acute phase of trauma, thereby intensifying coagulation disorders (14,15). Research from 2003 demonstrated that systemic coagulopathy can manifest within minutes following TBI (16), characterized by activation of protein C and enhanced fibrinolysis (17). Ordinarily, tissue-type plasminogen activator (tPA) struggles to access fibrin structures shielded by platelet aggregates, thus impeding the efficiency of subsequent enzymatic reactions essential for clot resolution (18). Moreover, the mechanical forces imparted on the brain during TBI mechanically disrupt the BBB in a pattern akin to a Gaussian distribution (19), precipitating secondary ischemic and inflammatory injuries. These injuries enhance the permeability of adjacent BBB segments, exacerbating the condition. Brain-derived microvesicles (BDMVs) enriched with tissue factor (TF) and phosphatidylserine (PS) are implicated in the exacerbation of brain injury, the initiation of early coagulation abnormalities and the promotion of hyperfibrinolysis (12). The elevated presence of TF and PS (12) in brain tissue not only facilitates the expansion of brain injury (20,21) but also instigates platelet dysfunction and depletion, as well as disseminated intravascular coagulation (DIC) (22,23). Lactoferrin, through its interaction with these microvesicles, has shown promise in mitigating coagulation disturbances and enhancing prognosis in a mouse model of TBI (24). Nonetheless, the specific mechanisms through which lactoferrin exerts its effects and the precise operational definitions of these cellular microvesicles continues to be elusive.

Neutrophils. Upon bodily injury, neutrophils are among the first cells to respond (25). Neutrophil extracellular traps (NETs), which are web-like structures released by activated neutrophils under specific conditions into the extracellular space, play a crucial role in entrapping and neutralizing microorganisms. The formation of NETs is a form of the programmed cell death process (26). Allen et al (27) have revealed that in the context of acute TBI, neutrophils are capable of penetrating cerebral vascular ECs and initiating the release of extracellular traps. This process triggers the release of procoagulant factors, including brain-derived microparticles and damage-associated molecular patterns (DAMPs) (27). The delayed apoptosis of neutrophils leads to an exacerbation of NETs formation and coagulation dysfunction during the acute phase of TBI (28). Platelet-neutrophil aggregation interactions play a pivotal role in traumatic coagulopathy. In patients with TBI who exhibit coagulation abnormalities, activated platelets enhance the formation of NETs generation through the secretion of high mobility group box 1 protein (HMGB1) (29,30). NETs exacerbate the disruption of the EC barrier by promoting phosphatidylserine exposure and TF expression on ECs (28,31,32). Moreover, NETs induce a procoagulant phenotype in ECs through the action of interleukin-1α and cathepsin G (33), which contributes to neurological impairment (34,35). Furthermore, inflammation plays a critical role in underlying secondary injury following TBI (36), with NETs significantly mediating the interaction between inflammation and coagulation (37). Given the comprehensive implications of these processes, NETs may offer a novel therapeutic targets for coagulopathy associated with TBI.

Platelets

Low platelet counts and/or functional platelet defects in platelets markedly enhance the risk of bleeding; a platelet count below 175x109/l is associated with an elevated risk of progressive intracranial hemorrhage progression, and counts below 100x109/l are strongly associated with increased mortality (38,39). This clinical presentation contrasts with traumatic coagulopathy (TIC), which is distinct from DIC, the latter typically characterized by thrombocytopenia (40). Firstly, in patients with TBI a reduced level of platelet reactivity is positively associated with improved prognoses (41,42). During TBI, the depletion of von Willebrand factor hampers platelet aggregation in vitro (43). This impairment is exacerbated by diminished platelet responsiveness to agonists such as adenosine diphosphate (ADP) and/or arachidonic acid (AA), leading to a specific defect in aggregation defects due to inhibition of ADP and AA receptors a phenomenon closely linked to TBI severity (22,44). These defects occur independently of hemorrhagic shock or the absolute platelet count (22,45). Additionally, elevated circulating levels of catecholamine platelet agonists, such as epinephrine and norepinephrine, are associated with compromised platelet aggregation function in patients with TBI (46).

This observation elucidates why even patients with mild injuries may exhibit suppressed platelet function in vitro (47). Variations in platelet activity may relate to systemic ischemia-reperfusion and oxidative stress (48), with declining platelet counts closely associated with injury severity and increased mortality risk (42,49). Overactive platelets may lead to secondary thrombocytopenia, heightening the risk of bleeding (22). Patients with TBI often exhibit moderately low platelet counts, with frequent activation of these cells, which generate microvesicles and display procoagulant activity (50,51). Secondly, platelet adhesion dysfunction is recognized as a pivotal factor in trauma response. Studies have revealed that in the aftermath of severe trauma, a notable reduction in in platelet adhesion to collagen is observed (52,53), along with diminished expression of specific receptors on platelets (54,55). These alterations may stem from fibrin-induced shedding of glycoprotein VI (GPVI) receptor (an important receptor on the surface of platelets that participates in collagen binding) (56) and interference with its signaling pathway (57).

Research conducted by Montague et al (58) has illustrated that inhibiting GPVI shedding by obstructing the fibrin-GPVI interaction offers a potential therapeutic avenue. Furthermore, GPVI shedding is predominantly facilitated by the metalloprotease ADAM10(59). In the context of trauma, platelets exhibit a distinctive ‘procoagulant’ phenotype, characterized by an unusual ‘swollen morphology’. This phenotype involves the translocation of PS from the inner to the outer leaflet of the cell membrane, thereby exposing PS on the platelet surface and creating a high-affinity substrate interface for thrombin generation (60). The prevalence of this morphological alteration correlates with injury severity, and these procoagulant platelets demonstrate compromised functionality in primary hemostasis (61). This indicates not only a diminished capacity for platelet aggregation in severe cases but also elevated markers of thrombin generation (62).

Collagen and thrombin, frequently found at sites of endothelial damage, conjointly stimulate the central mechanism for the development of procoagulant platelets in vitro, amplifying thrombin production (63). This process is accompanied by significant release of extracellular vesicles (EVs) (64). Certain studies have reported increased levels of platelet-derived EVs in plasma following major trauma, suggesting that purified platelet-derived EVs could potentially ameliorate thrombotic events post-trauma (65-68). Consequently, targeting the emergence of procoagulant platelets presents a promising therapeutic strategy for TIC.

3. Treatment of coagulation disorders following TBI

The early rectification of coagulopathy in patients with TBI is critically associated with survival rate (69). The predominant strategy for addressing TBI-induced coagulopathy entails the blood components, with a growing emphasis on the administration of clotting factors and related substances. Moreover, hemostatic agents are an essential element of the therapeutic arsenal employed in these cases, as their indispensable role in the management of coagulopathy post-TBI.

Pharmacological interventions

Tranexamic acid (TXA), a synthetic derivative of lysine, has proven effective in reducing active hemorrhage (70) and mortality (71) in trauma patients when administered within 3 h of injury, as demonstrated by the CRASH-2 trial (clinical randomization of an antifibrinolytic in significant hemorrhage) (71,72). Building on this, the CRASH-3 trial revealed that early administration of TXA within the same timeframe significantly reduces mortality risk in patients with mild-to-moderate TBI (67,73), thus endorsing its immediate use in such scenarios, whereas its efficacy diminishes in patients with severe TBI cases. Considering TXA's ability to enhance platelet function (74), coupled with its cost-effectiveness and proven efficacy in trauma management, it continues to be a dependable therapeutic option for patients with TBI.

Furthermore, desmopressin, as tested in a rat model of hemorrhagic shock, has been observed to elevate von Willebrand factor and factor VIII levels, as well as augment platelet aggregation (75). Given its short-acting nature and relative safety, desmopressin presents as a suitable alternative (76). Additional pharmacological agents such as progesterone, vitamin K2, Butylphthalide and recombinant interleukin-1 receptor antagonist (77) have demonstrated potential in the treatment of TBI. Conversely, administration of high-dose corticosteroids for 48 h to patients with moderate to severe brain injury has been associated with an increased mortality rate at two weeks (78). Amantadine may accelerate the rate of functional improvement in delirium-rating scale scores (79,80); however, due to its heterogeneity when used in patients with TBI (80), the benefits for this patient population requires further robust investigation. Although theoretically cytidine diphosphocholine (CDP-choline) might have positive effects on cell membrane integrity and cellular edema, studies indicate that its impact on cognitive function improvement in brain-injured patients appears to be negligible (81,82). Hemostatic agents can reduce bleeding in trauma patients, but excessive dosing or prolonged use may lead to acquired thrombosis (83). Therefore, when planning treatment, it is important to assess the risk and extent of thrombosis.

Transfusion of blood components following TBI

Post-TBI, blood component transfusions typically refer to the administration of platelets, red blood cells or plasma based on the individual needs of the patient. In scenarios of ongoing hemorrhage, platelet transfusions have not been proven effective in restoring aggregation function (84) nor have they demonstrated improvements in patient outcomes (85). Notably, the administration of refrigerated platelets has shown to confer superior hemostatic benefits compared with those stored at room temperature (86), a protocol now implemented in numerous trauma centers throughout the United States.

Furthermore, transfusions of packed red blood cells (pRBCs) have been recognized to enhance cerebral oxygenation; recent investigations into the combined administration of pRBCs with plasma in patients with TBI with coagulopathy have associated this practice with an escalation in adverse reactions and deteriorated prognoses (87). It is particularly noteworthy that establishing higher transfusion thresholds at 10 g/dl has been correlated with an upsurge in bleeding complications as opposed to a lower threshold of 7 g/dl (88), indicating that transfusion decisions should extend beyond mere adherence to rigid hemoglobin level. Consequently, a restrictive strategy for pRBC transfusion is advocated, except in instances where patients exhibit intolerance to anemia (89). The determination of the optimal timing and volume of transfusions remains a pivotal focus of ongoing research.

Hemostatic factors and associated substances

In recent years, advancements in the study of coagulation factors have significantly progressed the treatment of trauma patients. Fujiwara et al (90), employing a rat model of controlled cortical impact, demonstrated that daily intravenous injections of 350 mg/kg of a synthetically derived activated peptide of factor IX (termed F9-AP) significantly mitigated adjacent neuronal loss associated with secondary brain injury, markedly reducing both the volume of brain injury and associated edema. Moreover, recombinant activated factor VII, which exhibits lesser dependence on platelet function, appears to offer distinct advantages for patients with severely compromised platelet function or severe thrombocytopenia, effectively diminishing the risk of intracranial hemorrhage (91). Prothrombin complex concentrate (PCC), an inactivated blend comprising of factors II, IX, VII and X, has been proven to be highly efficacious in managing refractory bleeding to conventional treatment methods and in correcting elevated international normalized ratio (INR) levels (92). In TBI, where fibrinogen levels can be depleted, it is imperative to restore these levels to within normal ranges to alleviate inflammation and reduce endothelial permeability (93). The supplementation of factor XIII plays equally a critical role in inhibiting hyperfibrinolysis, stabilizing clot formations and minimizing surgical blood loss (94,95). However, compared with plasma, PCC may reduce hematoma expansion, yet shows no significant difference in 90-day mortality or Glasgow Coma Scale scores (96).

Reversal agents for direct oral anticoagulants (DOACs)

With an aging population, the number of patients with TBI concurrently taking oral anticoagulants is increasing. To ameliorate adverse outcomes including bleeding in patients undergoing surgery or those suffering from TBI while on these therapies, the development of antidotes for the reversal of DOACs has emerged as a crucial area of research. In 2015, publication of the first study demonstrating the safety and efficacy of idarucizumab, a targeted monoclonal antibody fragment for the acute reversal of dabigatran, a direct thrombin inhibitor, marked a significant advancement in anticoagulation management (97). This was followed by the introduction of Andexanet alfa, an antagonist of the facto Xa inhibitors, into clinical practice (98). Protamine sulfate is employed for the reversal of both unfractionated heparin and low molecular weight heparin (LMWH). However, its routine administration for reversing prophylactic subcutaneous heparin is not recommended unless there is a significantly prolonged activated partial thromboplastin time (aPTT) (99).

Notably, non-specific hemostatic agents such as PCC and activated prothrombin complex concentrate may also serve to reverse the effects of DOACs. However, FDA-approved reversal agents are not applicable to all DOACs or all clinical scenarios where reversal may be considered. Furthermore, the complexity of clinical use is compounded by factors such as cost, preparation and the lack of standardized protocols (97).

Potential therapies

Research underscores the complex and heterogeneous nature of TBI progression, necessitating exploration beyond conventional treatments toward interventions that may confer additional benefit to patients. This section delineates several promising potential therapeutic avenues post-TBI, aiming to foster groundbreaking advancements in the management during process.

It has been demonstrated that elevated levels of matrix metalloproteinase-9 (MMP-9) following brain injury contributes to the dysfunction of the BBB (100). Encouragingly, administration of MMP inhibitors in rodent models of brain injury has led to improved outcomes (101), indicating that MMP inhibition may represent a viable therapeutic strategy for patients with TBI. Moreover, the integration of medical science and nanotechnology has facilitated the development of platelet-mimicking nanovesicles, which incorporate the biological characteristics of platelet membranes into a lipid-based nanostructure. Through bioengineering manipulations, these particles can partially or fully emulate the hemostatic functions of natural platelets to varying extents (102), thus offering a promising alternative to platelet transfusion therapy. Additionally, novel resuscitative agents, known as platelet-derived extracellular vesicles (103), have surfaced, exhibiting remarkable hemostatic potency in patients with TBI via multiple mechanisms. However, current investigational agents remain in the experimental phase and additional research and data are required before they can advance to clinical application. As research advances and technology evolves, it is anticipated that additional mechanisms will be elucidated and applied to strategies aimed at ameliorating the outcomes for TBI survivors.

4. Complications

TBI can be accompanied by a wide array of complications, including but not limited to epilepsy, cerebral herniation, hydrocephalus and cerebrocardiac syndrome (104-106). These concomitant complications are intimately associated with a diminished quality of life and heightened mortality rates. Specifically, in the period following the initial day post-TBI, the emergence of multiple organ dysfunction and thrombotic events become the predominant causes of mortality among critically ill patients (107). Besides emphasizing early preventive strategies, it is equally essential to unravel the mechanisms that underlie the initiation and progression of these complications is equally indispensable. This underscores the imperative for a thorough comprehension and the formulation of precise intervention strategies.

Multiple organ dysfunction syndrome (MODS)

Post-traumatic MODS is widely recognized as a consequence of deregulated responses to trauma. The primary pathogenic mechanism likely involves the activation of both coagulation and inflammatory cascades (108,109). Furthermore, MODS is intimately associated with extracellular histones, HMGB1 and S100A8/9, among other factors. Notably, elevated levels of HMGB1 contribute to organ injury (109). Additionally, the dynamic interaction between platelets and leukocytes facilitates leukocyte recruitment to sites of injury, facilitating tissue repair (110). Nevertheless, an excessive immune response appears to inflict organ damage (111). Fortunately, proactive management of coagulopathy exhibits promising potential in ameliorating the incidence of organ failure (112).

Thrombosis

Thromboembolic events play a significant cause in increasing morbidity and disability rates among patients, particularly those who have suffered TBI and are severely injured or unable to mobilize independently. Considering TBI as an established independent risk factor for venous thromboembolism (VTE) (113,114). The present discussion predominantly focuses on VTE, extending beyond traditional triggers to explore the pivotal role played by the interaction between platelets and monocytes in the initiation and propagation of VTE (115). Moreover, factors including NETs, platelet-derived microparticles and protein C depletion (38) are intimately associated with VTE development. In intensive care units, the occurrence of VTE among critically ill trauma patients is notably elevated, with an estimated incidence of up to 35% (116). Even with the implementation of mechanical prophylaxis measures for deep vein thrombosis (DVT), residual DVT and pulmonary embolism (PE) rates remain substantial at 31 and 3% (114), respectively. A recent large comprehensive observational study emphasized that de novo pulmonary thrombi are more prevalent than PEs originating from DVT, often manifesting early in the clinical course (117). In animal models of TBI, microthrombi are predominantly observed in the pericontusional cortex (118), composed of fibrin, platelets and other components. LMWH has demonstrated superior efficacy in preventing thromboembolism compared to unfractionated heparin (119), indicating that, following a thorough patient evaluation and confirmation of no contraindications, early initiation of LMWH for thromboprophylaxis is recommended. Apart from LMWH, factor XI inhibitors, such as Abelacimab, have shown effectiveness in preventing VTE (120), offering an alternative preventive strategy in managing thrombotic risks.

Consequently, early detection, prevention and management of MODS and thrombotic events assume paramount importance. Identification and assessment of disease severity, as well as prediction of risks in patients, are facilitated through monitoring vital signs, imaging changes, laboratory parameters and scoring systems such as APACHE II, SOFA and qSOFA (121,122). Management of these patients should be viewed as a dynamic process, necessitating close surveillance and prompt adjustment of therapeutic strategies as needed. Given the unique nature of each patient's condition, treatment plans should be individualized, with the overarching goals of maximizing organ function recovery, preventing thrombosis and enhancing quality of life.

5. Diagnostic tools for coagulopathy following TBI

Laboratory assessments aimed at detecting coagulopathy and techniques reflecting brain injury are crucial in diagnosing and managing coagulation disorders that arise following TBI (Table II). Conventional coagulation assays, routinely employed in clinical settings to evaluate hemostatic function, encompass prothrombin time, aPTT and INR (123,124), which aid in prognostication post-cranial injury. Nonetheless, these tests fail to capture the full complexity of coagulation processes and have limitations in accurately representing thrombin generation and precision in hemostatic evaluation (125,126).

Table II

Advantages and disadvantages of various diagnostic tools.

Table II

Advantages and disadvantages of various diagnostic tools.

Diagnostic toolsContentAdvantagesDisadvantagesClinical application
CCAsTests such as prothrombin time, activated partial thromboplastin time and international normalized ratioIt can indirectly predict the prognosis of patients with coagulopathyIt is unable to provide real-time information on coagulation changes; it does not assess the levels of thrombin or the extent of clot formation involved in the reactionConventional methods
ROTEMDetailed kinetics of blood coagulation such as clot initiation time, fibrin formation velocity, clot firmness and fibrinolysis activityIt allows for real-time assessment of hemostatic status, rapid feedback on test results and prediction of treatment efficacy; it can quickly and accurately evaluate fibrinogen levels; and it can guide therapeutic interventionsIt requires stringent technical expertise; interpretation of results still relies heavily on the operator's experience, with lower standardization compared to conventional tests; and it is relatively more costlyPreferred methods
TEGReaction time; kinetic time; alpha angle; maximum amplitude; lysis at 30 minIt allows for real-time assessment of hemostatic status, rapid feedback on test results and prediction of treatment efficacy; it can quickly and accurately evaluate fibrinogen levels; and it can guide therapeutic interventionsIt demands strict technical requirements; results can be influenced by human factors; and it is time-consumingPreferred methods
POC-PFTTEG (TEG 5000/6 s Hemostasis Analyzer System), ROTEM (ROTEM Delta System), platelet reactivity turbidimetry test (VerifyNow System), multiple electrode platelet aggregometry (Multiplate Analyzer) and PFA (PFA-100/200)It enables real-time monitoring of platelet function: Reducing blood transfusion in cardiac surgery and in predicting the risk of perioperative blood loss in high risk cardiac surgery patientsThere is no universally recognized ‘gold standard’; it requires stringent technical expertise; and the results can be subject to human-related variablesNot routinely utilized

[i] ROTEM, rotational thromboelastography; TEG, thromboelastography; PFA, platelet function analyzer.

More advanced global hemostasis assessments, including rotational thromboelastometry (ROTEM), thromboelastography (TEG) and thrombin generation tests, may offer a superior real-time analysis of hemostatic status. These provide swift feedback for therapeutic intervention and enable more precise predictions treatment outcomes (127). These methodologies further facilitate goal-directed transfusion strategies (128) and guide heparin administration for thromboembolism prophylaxis (129), with TEG particularly unaffected by the administration of TXA (130). A fibrinogen level concentration 2.0 g/l is recognized as a risk factor for coagulopathy and associated complications post-TBI. Both ROTEM and TEG have proven effective in swiftly and precisely measuring fibrinogen levels (127). Moreover, recent research involving bleeding adult and pediatric patients has demonstrated that transfusion strategies guided by viscoelastic tests improve survival rates, decrease blood product usage and reduce the incidence of renal failure when compared with other methods (131). Point-of-care platelet function testing (POC-PFT), exemplified by systems including VerifyNow, Plateletworks, PFA-100/200, show promise in identifying platelet dysfunction or guiding antiplatelet therapy (6,20). However, the absence of a gold standard for POC-PFT and substantial variation among available analyzers in terms of implementation technique and platelet agonists utilized hinder widespread acceptance. Consequently, current European guidelines on massive hemorrhage and coagulation management in trauma do not advocate for the routine use of POC-PFT (132).

6. Neurocritical care monitoring tools

For patients suffering from moderate to severe brain injuries, the mere reliance on conventional blood tests is insufficient, Conventional coagulation tests measure only ~4% of total thrombin generation and do not assess the overall hemostatic state. Furthermore, these tests do not reflect the interactions between multiple coagulation pathway mechanisms (133-135). Therefore, the incorporation of cutting-edge neurocritical care monitoring instruments is of utmost importance. In this context, multimodal monitoring (MMM) encompasses a range of techniques, including intracranial pressure (ICP)-monitoring, cerebral microdialysis (CMD), cerebral tissue oxygenation monitoring and continuous electroencephalography (EEG) (3), amongst others. Through the amalgamation of these diverse monitoring modalities, MMM provides a comprehensive, real-time evaluation of the patient's cerebral pathophysiology. This significantly bolsters the management of brain disorders, elevates patient outcomes and propels clinical research forward.

Primarily, ICP monitoring stands as a pivotal component, as elevated ICP can diminish cerebral perfusion [cerebral perfusion pressure=mean arterial pressure (MAP)-ICP], subsequently augmenting the peril of ischemia and herniation (136,137). In accordance with the established guidelines, maintaining ICP at 20-25 mmHg (138), with MAP kept at 60-70 mmHg in severe patients with TBI, is paramount in effectively mitigating adverse outcomes. Furthermore, CMD emerges as an invasive yet invaluable technique. It involves hourly sampling and analysis of cerebral extracellular fluid metabolites, offering an unprecedented glimpse into the biochemical shifts within the brain (139). Complementing this, cerebral tissue oxygenation monitoring employs near-infrared spectroscopy for a non-invasive, continuous assessment of regional concentrations of oxygenated and deoxygenated hemoglobin in the brain (140). This, in turn, aids in evaluating the risk of cerebral hypoxia and shaping individualized therapeutic strategies. However, it's worth noting that its application remains primarily confined to superficial areas such as the frontal cortex, owing to technological limitations.

In the realm of severe TBI, post-traumatic seizures affect roughly one in ten patients, with asymptomatic seizure activity prevalence potentially escalating to 20-25% (141). Continuous EEG enables the early detection of these cerebral pathologies and guides antiepileptic therapy. As previously reported, the successful management of a 25-year-old comatose patient following a car accident was achieved through the utilization of the MMM monitoring and management system. This case underscores the practical utility of MMM. The system is capable of providing real-time updates on physiological changes and offering early indications of potential issues before they escalate, thereby guiding preemptive interventions (142). In conclusion, with the progression of technology and the accumulation of clinical experience, the application of MMM is likely to evolve into a more refined and widespread practice. IF so, it would be an indispensable component of modern neurointensive care.

7. Summary and perspectives

TBI triggers swift and substantial modifications in cellular behaviors, thereby amplifying the intricacy of coagulopathy mechanisms and complicating treatment options by altering both intra- and intercellular connections. The diverse changes induced by TBI are still inadequately targeted by current therapeutic approaches. As a result, there is an urgent necessity for deeper investigation to clarify the fundamental mechanisms underlying coagulopathy of post-TBI and to refine precision therapies accordingly. This involves establishing diagnostic thresholds and developing targeted therapeutic interventions aimed at effectively managing coagulation disorders. Current research lacks prospective diagnostic modalities that can accurately determine the timepoint of hemostatic failure, making it challenging to define the optimal timing for initiating anticoagulation therapy in patients with coagulopathy. The aging population of patients with TBI, coupled with the widespread use of antiplatelet medications and anticoagulants, poses a considerable challenge in comprehending the distinct impacts of various antiplatelet medications and anticoagulants on post-TBI coagulopathy. Tackling these challenges is paramount for improving clinical outcomes and minimizing the morbidity and mortality associated with this condition. Hence, there is a critical need for more expeditious and efficacious techniques to restore coagulation, thereby limiting the progression of brain injury in patients on anticoagulant and antiplatelet medications, with the ultimate aim of improving outcomes. Moreover, future research focusing on the management of coagulopathies in both critically and non-critically ill patients post-TBI should prioritize not just survival rates, but also enhancements in quality of life for patients.

Acknowledgements

The authors are grateful to Ms. Du Pengchao (Binzhou Medical College, Yantai, China) for her insightful suggestions and constructive feedback on the manuscript.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

HH and ZQ performed the literature search and co-wrote the manuscript. RL, BJ, LW and AL participated in the literature search and reviewed the literature and the manuscript. HH, ZQ, RL, BJ, LW and AL made substantial contributions to the conception and revision of the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tierney RT, Mansell JL, Higgins M, McDevitt JK, Toone N, Gaughan JP, Mishra A and Krynetskiy E: Apolipoprotein E genotype and concussion in college athletes. Clin J Sport Med. 20:464–468. 2010.PubMed/NCBI View Article : Google Scholar

2 

Panenka WJ, Gardner AJ, Dretsch MN, Crynen GC, Crawford FC and Iverson GL: Systematic review of genetic risk factors for sustaining a mild traumatic brain injury. J Neurotrauma. 34:2093–2099. 2017.PubMed/NCBI View Article : Google Scholar

3 

Khellaf A, Khan DZ and Helmy A: Recent advances in traumatic brain injury. J Neurol. 266:2878–2889. 2019.PubMed/NCBI View Article : Google Scholar

4 

Maegele M, Aversa J, Marsee MK, McCauley R, Chitta SH, Vyakaranam S and Walsh M: Changes in coagulation following brain injury. Semin Thromb Hemost. 46:155–166. 2020.PubMed/NCBI View Article : Google Scholar

5 

Roozenbeek B, Maas AI and Menon DK: Changing patterns in the epidemiology of traumatic brain injury. Nat Rev Neurol. 9:231–236. 2013.PubMed/NCBI View Article : Google Scholar

6 

Prinz V, Finger T, Bayerl S, Rosenthal C, Wolf S, Liman T and Vajkoczy P: High prevalence of pharmacologically induced platelet dysfunction in the acute setting of brain injury. Acta Neurochir (Wien). 158:117–123. 2016.PubMed/NCBI View Article : Google Scholar

7 

Shehata M, Afify M, El-Shafie M and Khaled M: Prevalence and clinical implications of coagulopathy in patients with isolated head trauma. Med J Cairo Univ. 79:131–137. 2011.

8 

Harhangi BS, Kompanje EJ, Leebeek FW and Maas AI: Coagulation disorders after traumatic brain injury. Acta Neurochir (Wien). 150:165–175; discussion 175. 2008.PubMed/NCBI View Article : Google Scholar

9 

Hartings JA, Bullock MR, Okonkwo DO, Murray LS, Murray GD, Fabricius M, Maas AI, Woitzik J, Sakowitz O, Mathern B, et al: Spreading depolarisations and outcome after traumatic brain injury: A prospective observational study. Lancet Neurol. 10:1058–1064. 2011.PubMed/NCBI View Article : Google Scholar

10 

Mena JH, Sanchez AI, Rubiano AM, Peitzman AB, Sperry JL, Gutierrez MI and Puyana JC: Effect of the modified Glasgow Coma Scale score criteria for mild traumatic brain injury on mortality prediction: Comparing classic and modified Glasgow Coma Scale score model scores of 13. J Trauma. 71:1185–1192; discussion 1193. 2011.PubMed/NCBI View Article : Google Scholar

11 

Maegele M, Schöchl H, Menovsky T, Maréchal H, Marklund N, Buki A and Stanworth S: Coagulopathy and haemorrhagic progression in traumatic brain injury: Advances in mechanisms, diagnosis, and management. Lancet Neurol. 16:630–647. 2017.PubMed/NCBI View Article : Google Scholar

12 

Tian Y, Salsbery B, Wang M, Yuan H, Yang J, Zhao Z, Wu X, Zhang Y, Konkle BA, Thiagarajan P, et al: Brain-derived microparticles induce systemic coagulation in a murine model of traumatic brain injury. Blood. 125:2151–2159. 2015.PubMed/NCBI View Article : Google Scholar

13 

Zhao Z, Wang M, Tian Y, Hilton T, Salsbery B, Zhou EZ, Wu X, Thiagarajan P, Boilard E, Li M, et al: Cardiolipin-mediated procoagulant activity of mitochondria contributes to traumatic brain injury–associated coagulopathy in mice. Blood. 127:2763–2772. 2016.PubMed/NCBI View Article : Google Scholar

14 

Albert V, Subramanian A, Agrawal D, Pati HP, Gupta SD and Mukhopadhyay AK: Acute Traumatic endotheliopathy in isolated severe brain injury and its impact on clinical outcome. Med Sci (Basel). 6(5)2018.PubMed/NCBI View Article : Google Scholar

15 

Sillesen M, Rasmussen LS, Jin G, Jepsen CH, Imam A, Hwabejire JO, Halaweish I, DeMoya M, Velmahos G, Johansson PI and Alam HB: Assessment of coagulopathy, endothelial injury, and inflammation after traumatic brain injury and hemorrhage in a porcine model. J Trauma Acute Care Surg. 76:12–20. 2014.PubMed/NCBI View Article : Google Scholar

16 

Maegele M: Coagulopathy after traumatic brain injury: Incidence, pathogenesis, and treatment options. Transfusion. 53 (Suppl 1):28S–37S. 2013.PubMed/NCBI View Article : Google Scholar

17 

Hulka F, Mullins RJ and Frank EH: Blunt brain injury activates the coagulation process. Arch Surg. 131:923–928. 1996.PubMed/NCBI View Article : Google Scholar

18 

Brommer EJ: The level of extrinsic plasminogen activator (t-PA) during clotting as a determinant of the rate of fibrinolysis; Inefficiency of activators added afterwards. Thromb Res. 34:109–115. 1984.PubMed/NCBI View Article : Google Scholar

19 

Haltmeier T, Benjamin E, Gruen JP, Shulman IA, Lam L, Inaba K and Demetriades D: Decreased mortality in patients with isolated severe blunt traumatic brain injury receiving higher plasma to packed red blood cells transfusion ratios. Injury. 49:62–66. 2018.PubMed/NCBI View Article : Google Scholar

20 

Fleck RA, Rao LV, Rapaport SI and Varki N: Localization of human tissue factor antigen by immunostaining with monospecific, polyclonal anti-human tissue factor antibody. Thromb Res. 59:421–437. 1990.PubMed/NCBI View Article : Google Scholar

21 

Eddleston M, de la Torre JC, Oldstone MB, Loskutoff DJ, Edgington TS and Mackman N: Astrocytes are the primary source of tissue factor in the murine central nervous system. A role for astrocytes in cerebral hemostasis. J Clin Invest. 92:349–358. 1993.PubMed/NCBI View Article : Google Scholar

22 

Castellino FJ, Chapman MP, Donahue DL, Thomas S, Moore EE, Wohlauer MV, Fritz B, Yount R, Ploplis V, Davis P, et al: Traumatic brain injury causes platelet adenosine diphosphate and arachidonic acid receptor inhibition independent of hemorrhagic shock in humans and rats. J Trauma Acute Care Surg. 76:1169–1176. 2014.PubMed/NCBI View Article : Google Scholar

23 

Hoffman M and Monroe DM: Tissue factor in brain is not saturated with factor VIIa: Implications for factor VIIa dosing in intracerebral hemorrhage. Stroke. 40:2882–2884. 2009.PubMed/NCBI View Article : Google Scholar

24 

Zhou Y, Cai W, Zhao Z, Hilton T, Wang M, Yeon J, Liu W, Zhang F, Shi FD, Wu X, et al: Lactadherin promotes microvesicle clearance to prevent coagulopathy and improves survival of severe TBI mice. Blood. 131:563–572. 2018.PubMed/NCBI View Article : Google Scholar

25 

Kolaczkowska E and Kubes P: Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 13:159–175. 2013.PubMed/NCBI View Article : Google Scholar

26 

Sollberger G, Tilley DO and Zychlinsky A: Neutrophil extracellular traps: The biology of chromatin externalization. Dev Cell. 44:542–553. 2018.PubMed/NCBI View Article : Google Scholar

27 

Allen C, Thornton P, Denes A, McColl BW, Pierozynski A, Monestier M, Pinteaux E, Rothwell NJ and Allan SM: Neutrophil cerebrovascular transmigration triggers rapid neurotoxicity through release of proteases associated with decondensed DNA. J Immunol. 189:381–392. 2012.PubMed/NCBI View Article : Google Scholar

28 

Jin J, Wang F, Tian J, Zhao X, Dong J, Wang N, Liu Z, Zhao H, Li W, Mang G and Hu S: Neutrophil extracellular traps contribute to coagulopathy after traumatic brain injury. JCI Insight. 8(e141110)2023.PubMed/NCBI View Article : Google Scholar

29 

Maugeri N, Capobianco A, Rovere-Querini P, Ramirez GA, Tombetti E, Valle PD, Monno A, D'Alberti V, Gasparri AM, Franchini S, et al: Platelet microparticles sustain autophagy-associated activation of neutrophils in systemic sclerosis. Sci Transl Med. 10(eaao3089)2018.PubMed/NCBI View Article : Google Scholar

30 

Jiao Y, Li W, Wang W, Tong X, Xia R, Fan J, Du J, Zhang C and Shi X: Platelet-derived exosomes promote neutrophil extracellular trap formation during septic shock. Crit Care. 24(380)2020.PubMed/NCBI View Article : Google Scholar

31 

Frangou E, Chrysanthopoulou A, Mitsios A, Kambas K, Arelaki S, Angelidou I, Arampatzioglou A, Gakiopoulou H, Bertsias GK, Verginis P, et al: REDD1/autophagy pathway promotes thromboinflammation and fibrosis in human systemic lupus erythematosus (SLE) through NETs decorated with tissue factor (TF) and interleukin-17A (IL-17A). Ann Rheum Dis. 78:238–248. 2019.PubMed/NCBI View Article : Google Scholar

32 

Stakos DA, Kambas K, Konstantinidis T, Mitroulis I, Apostolidou E, Arelaki S, Tsironidou V, Giatromanolaki A, Skendros P, Konstantinides S and Ritis K: Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur Heart J. 36:1405–1414. 2015.PubMed/NCBI View Article : Google Scholar

33 

Folco EJ, Mawson TL, Vromman A, Bernardes-Souza B, Franck G, Persson O, Nakamura M, Newton G, Luscinskas FW and Libby P: Neutrophil extracellular traps induce endothelial cell activation and tissue factor production through Interleukin-1α and Cathepsin G. Arterioscler Thromb Vasc Biol. 38:1901–1912. 2018.PubMed/NCBI View Article : Google Scholar

34 

Zhu K, Zhu Y, Hou X, Chen W, Qu X, Zhang Y, Li Z, Wang C, Chen J, Lv L, et al: NETs Lead to sympathetic hyperactivity after traumatic brain injury through the LL37-Hippo/MST1 Pathway. Front Neurosci. 15(621477)2021.PubMed/NCBI View Article : Google Scholar

35 

Vaibhav K, Braun M, Alverson K, Khodadadi H, Kutiyanawalla A, Ward A, Banerjee C, Sparks T, Malik A, Rashid MH, et al: Neutrophil extracellular traps exacerbate neurological deficits after traumatic brain injury. Sci Adv. 6(eaax8847)2020.PubMed/NCBI View Article : Google Scholar

36 

Jassam YN, Izzy S, Whalen M, McGavern DB and El Khoury J: Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron. 95:1246–1265. 2017.PubMed/NCBI View Article : Google Scholar

37 

Noubouossie DF, Reeves BN, Strahl BD and Key NS: Neutrophils: Back in the thrombosis spotlight. Blood. 133:2186–2197. 2019.PubMed/NCBI View Article : Google Scholar

38 

Laroche M, Kutcher ME, Huang MC, Cohen MJ and Manley GT: Coagulopathy after traumatic brain injury. Neurosurgery. 70:1334–1345. 2012.PubMed/NCBI View Article : Google Scholar

39 

Joseph B, Aziz H, Zangbar B, Kulvatunyou N, Pandit V, O'Keeffe T, Tang A, Wynne J, Friese RS and Rhee P: Acquired coagulopathy of traumatic brain injury defined by routine laboratory tests: Which laboratory values matter? J Trauma Acute Care Surg. 76:121–125. 2014.PubMed/NCBI View Article : Google Scholar

40 

Wada H, Thachil J, Di Nisio M, Mathew P, Kurosawa S, Gando S, Kim HK, Nielsen JD, Dempfle CE, Levi M, et al: Guidance for diagnosis and treatment of DIC from harmonization of the recommendations from three guidelines. J Thromb Haemost: Feb 4, 2013 (Epub ahead of print).

41 

McCully BH, Connelly CR, Fair KA, Holcomb JB, Fox EE, Wade CE, Bulger EM and Schreiber MA: PROPPR Study Group. Onset of coagulation function recovery is delayed in severely injured trauma patients with venous thromboembolism. J Am Coll Surg. 225:42–51. 2017.PubMed/NCBI View Article : Google Scholar

42 

Solomon C, Traintinger S, Ziegler B, Hanke A, Rahe-Meyer N, Voelckel W and Schöchl H: Platelet function following trauma. A multiple electrode aggregometry study. Thromb Haemost. 106:322–330. 2011.PubMed/NCBI View Article : Google Scholar

43 

Kornblith LZ, Robles AJ, Conroy AS, Hendrickson CM, Calfee CS, Fields AT, Callcut RA and Cohen MJ: Perhaps it's not the platelet: Ristocetin uncovers the potential role of von Willebrand factor in impaired platelet aggregation following traumatic brain injury. J Trauma Acute Care Surg. 85:873–880. 2018.PubMed/NCBI View Article : Google Scholar

44 

Davis PK, Musunuru H, Walsh M, Cassady R, Yount R, Losiniecki A, Moore EE, Wohlauer MV, Howard J, Ploplis VA, et al: Platelet dysfunction is an early marker for traumatic brain injury-induced coagulopathy. Neurocrit Care. 18:201–208. 2013.PubMed/NCBI View Article : Google Scholar

45 

Briggs A, Gates JD, Kaufman RM, Calahan C, Gormley WB and Havens JM: Platelet dysfunction and platelet transfusion in traumatic brain injury. J Surg Res. 193:802–806. 2015.PubMed/NCBI View Article : Google Scholar

46 

Matthay ZA, Fields AT, Nunez-Garcia B, Park JJ, Jones C, Leligdowicz A, Hendrickson CM, Callcut RA, Matthay MA and Kornblith LZ: Importance of catecholamine signaling in the development of platelet exhaustion after traumatic injury. J Thromb Haemost. 20:2109–2118. 2022.PubMed/NCBI View Article : Google Scholar

47 

Sirajuddin S, Valdez C, DePalma L, Maluso P, Singhal R, Schroeder M and Sarani B: Inhibition of platelet function is common following even minor injury. J Trauma Acute Care Surg. 81:328–332. 2016.PubMed/NCBI View Article : Google Scholar

48 

Kutcher ME, Redick BJ, McCreery RC, Crane IM, Greenberg MD, Cachola LM, Nelson MF and Cohen MJ: Characterization of platelet dysfunction after trauma. J Trauma Acute Care Surg. 73:13–19. 2012.PubMed/NCBI View Article : Google Scholar

49 

Brown LM, Call MS, Margaret Knudson M and Cohen MJ: Trauma Outcomes Group. Holcomb JB, Wade CE, Brasel KJ, Vercruysse G, MacLeod J, et al: A normal platelet count may not be enough: The impact of admission platelet count on mortality and transfusion in severely injured trauma patients. J Trauma. 71 (2 Suppl 3):S337–S342. 2011.PubMed/NCBI View Article : Google Scholar

50 

Ploplis VA, Donahue DL, Sandoval-Cooper MJ, MorenoCaffaro M, Sheets P, Thomas SG, Walsh M and Castellino FJ: Systemic platelet dysfunction is the result of local dysregulated coagulation and platelet activation in the brain in a rat model of isolated traumatic brain injury. J Neurotrauma. 31:1672–1675. 2014.PubMed/NCBI View Article : Google Scholar

51 

Prodan CI, Vincent AS and Dale GL: Coated-Platelet levels increase with number of injuries in patients with mild traumatic brain injury. J Neurotrauma. 33:818–824. 2016.PubMed/NCBI View Article : Google Scholar

52 

Vulliamy P, Montague SJ, Gillespie S, Chan MV, Coupland LA, Andrews RK, Warner TD, Gardiner EE, Brohi K and Armstrong PC: Loss of GPVI and GPIbα contributes to trauma-induced platelet dysfunction in severely injured patients. Blood Adv. 4:2623–2630. 2020.PubMed/NCBI View Article : Google Scholar

53 

Li R, Elmongy H, Sims C and Diamond SL: Ex vivo recapitulation of trauma-induced coagulopathy and preliminary assessment of trauma patient platelet function under flow using microfluidic technology. J Trauma Acute Care Surg. 80:440–449. 2016.PubMed/NCBI View Article : Google Scholar

54 

Fields AT, Matthay ZA, Nunez-Garcia B, Matthay EC, Bainton RJ, Callcut RA and Kornblith LZ: Good platelets gone bad: The effects of trauma patient plasma on healthy platelet aggregation. Shock. 55:189–197. 2021.PubMed/NCBI View Article : Google Scholar

55 

Verni CC, Davila A Jr, Balian S, Sims CA and Diamond SL: Platelet dysfunction during trauma involves diverse signaling pathways and an inhibitory activity in patient-derived plasma. J Trauma Acute Care Surg. 86:250–259. 2019.PubMed/NCBI View Article : Google Scholar

56 

Montague SJ, Delierneux C, Lecut C, Layios N, Dinsdale RJ, Lee CS, Poulter NS, Andrews RK, Hampson P, Wearn CM, et al: Soluble GPVI is elevated in injured patients: shedding is mediated by fibrin activation of GPVI. Blood Adv. 2:240–251. 2018.PubMed/NCBI View Article : Google Scholar

57 

Lee MY, Verni CC, Herbig BA and Diamond SL: Soluble fibrin causes an acquired platelet glycoprotein VI signaling defect: Implications for coagulopathy. J Thromb Haemost. 15:2396–2407. 2017.PubMed/NCBI View Article : Google Scholar

58 

Montague SJ, Hicks SM, Lee CS, Coupland LA, Parish CR, Lee WM, Andrews RK and Gardiner EE: Fibrin exposure triggers αIIbβ3-independent platelet aggregate formation, ADAM10 activity and glycoprotein VI shedding in a charge-dependent manner. J Thromb Haemost. 18:1447–1458. 2020.PubMed/NCBI View Article : Google Scholar

59 

Gardiner EE, Karunakaran D, Shen Y, Arthur JF, Andrews RK and Berndt MC: Controlled shedding of platelet glycoprotein (GP)VI and GPIb-IX-V by ADAM family metalloproteinases. J Thromb Haemost. 5:1530–1537. 2007.PubMed/NCBI View Article : Google Scholar

60 

Agbani EO, van den Bosch MT, Brown E, Williams CM, Mattheij NJ, Cosemans JM, Collins PW, Heemskerk JW, Hers I and Poole AW: Coordinated membrane ballooning and procoagulant spreading in human platelets. Circulation. 132:1414–1424. 2015.PubMed/NCBI View Article : Google Scholar

61 

Kulkarni S, Woollard KJ, Thomas S, Oxley D and Jackson SP: Conversion of platelets from a proaggregatory to a proinflammatory adhesive phenotype: Role of PAF in spatially regulating neutrophil adhesion and spreading. Blood. 110:1879–1886. 2007.PubMed/NCBI View Article : Google Scholar

62 

Vulliamy P, Gillespie S, Armstrong PC, Allan HE, Warner TD and Brohi K: Histone H4 induces platelet ballooning and microparticle release during trauma hemorrhage. Proc Natl Acad Sci USA. 116:17444–17449. 2019.PubMed/NCBI View Article : Google Scholar

63 

Agbani EO and Poole AW: Procoagulant platelets: Generation, function, and therapeutic targeting in thrombosis. Blood. 130:2171–2179. 2017.PubMed/NCBI View Article : Google Scholar

64 

Boilard E, Duchez AC and Brisson A: The diversity of platelet microparticles. Curr Opin Hematol. 22:437–444. 2015.PubMed/NCBI View Article : Google Scholar

65 

Curry N, Raja A, Beavis J, Stanworth S and Harrison P: Levels of procoagulant microvesicles are elevated after traumatic injury and platelet microvesicles are negatively correlated with mortality. J Extracell Vesicles. 3(25625)2014.PubMed/NCBI View Article : Google Scholar

66 

Matijevic N, Wang YW, Holcomb JB, Kozar R, Cardenas JC and Wade CE: Microvesicle phenotypes are associated with transfusion requirements and mortality in subjects with severe injuries. J Extracell Vesicles. 4(29338)2015.PubMed/NCBI View Article : Google Scholar

67 

Lopez E, Srivastava AK, Pati S, Holcomb JB and Wade CE: Platelet-Derived microvesicles: A potential therapy for trauma-induced coagulopathy. Shock. 49:243–248. 2018.PubMed/NCBI View Article : Google Scholar

68 

Price J, Gardiner C and Harrison P: Platelet-enhanced plasma: Characterization of a novel candidate resuscitation fluid's extracellular vesicle content, clotting parameters, and thrombin generation capacity. Transfusion. 61:2179–2194. 2021.PubMed/NCBI View Article : Google Scholar

69 

Epstein DS, Mitra B, Cameron PA, Fitzgerald M and Rosenfeld JV: Normalization of coagulopathy is associated with improved outcome after isolated traumatic brain injury. J Clin Neurosci. 29:64–69. 2016.PubMed/NCBI View Article : Google Scholar

70 

Dewan Y, Komolafe EO, Mejía-Mantilla JH, Perel P, Roberts I and Shakur H: CRASH-3 Collaborators. CRASH-3-tranexamic acid for the treatment of significant traumatic brain injury: study protocol for an international randomized, double-blind, placebo-controlled trial. Trials. 13(87)2012.PubMed/NCBI View Article : Google Scholar

71 

CRASH-2 trial collaborators. Shakur H, Roberts I, Bautista R, Caballero J, Coats T, Dewan Y, El-Sayed H, Gogichaishvili T, Gupta S, et al: Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): A randomised, placebo-controlled trial. Lancet. 376:23–32. 2010.PubMed/NCBI View Article : Google Scholar

72 

Roberts I, Shakur H, Coats T, Hunt B, Balogun E, Barnetson L, Cook L, Kawahara T, Perel P, Prieto-Merino D, et al: The CRASH-2 trial: A randomised controlled trial and economic evaluation of the effects of tranexamic acid on death, vascular occlusive events and transfusion requirement in bleeding trauma patients. Health Technol Assess. 17:1–79. 2013.PubMed/NCBI View Article : Google Scholar

73 

CRASH-3 trial collaborators. Effects of tranexamic acid on death, disability, vascular occlusive events and other morbidities in patients with acute traumatic brain injury (CRASH-3): A randomised, placebo-controlled trial. Lancet. 394:1713–1723. 2019.PubMed/NCBI View Article : Google Scholar

74 

Weber CF, Görlinger K, Byhahn C, Moritz A, Hanke AA, Zacharowski K and Meininger D: Tranexamic acid partially improves platelet function in patients treated with dual antiplatelet therapy. Eur J Anaesthesiol. 28:57–62. 2011.PubMed/NCBI View Article : Google Scholar

75 

Naidech AM, Maas MB, Levasseur-Franklin KE, Liotta EM, Guth JC, Berman M, Rosenow JM, Lindholm PF, Bendok BR, Prabhakaran S, et al: Desmopressin improves platelet activity in acute intracerebral hemorrhage. Stroke. 45:2451–2453. 2014.PubMed/NCBI View Article : Google Scholar

76 

Kim DY, O'Leary M, Nguyen A, Kaji A, Bricker S, Neville A, Bongard F, Putnam B and Plurad D: The effect of platelet and desmopressin administration on early radiographic progression of traumatic intracranial hemorrhage. J Neurotrauma. 32:1815–1821. 2015.PubMed/NCBI View Article : Google Scholar

77 

Helmy A, Guilfoyle MR, Carpenter KL, Pickard JD, Menon DK and Hutchinson PJ: Recombinant human interleukin-1 receptor antagonist in severe traumatic brain injury: A phase II randomized control trial. J Cereb Blood Flow Metab. 34:845–851. 2014.PubMed/NCBI View Article : Google Scholar

78 

Roberts I, Yates D, Sandercock P, Farrell B, Wasserberg J, Lomas G, Cottingham R, Svoboda P, Brayley N, Mazairac G, et al: Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury (MRC CRASH trial): Randomised placebo-controlled trial. Lancet. 364:1321–1328. 2004.PubMed/NCBI View Article : Google Scholar

79 

Mizoguchi K, Yokoo H, Yoshida M, Tanaka T and Tanaka M: Amantadine increases the extracellular dopamine levels in the striatum by re-uptake inhibition and by N-methyl-D-aspartate antagonism. Brain Res. 662:255–258. 1994.PubMed/NCBI View Article : Google Scholar

80 

Giacino JT, Whyte J, Bagiella E, Kalmar K, Childs N, Khademi A, Eifert B, Long D, Katz DI, Cho S, et al: Placebo-controlled trial of amantadine for severe traumatic brain injury. N Engl J Med. 366:819–826. 2012.PubMed/NCBI View Article : Google Scholar

81 

Secades JJ: Role of citicoline in the management of traumatic brain injury. Pharmaceuticals (Basel). 14(410)2021.PubMed/NCBI View Article : Google Scholar

82 

Zafonte RD, Bagiella E, Ansel BM, Novack TA, Friedewald WT, Hesdorffer DC, Timmons SD, Jallo J, Eisenberg H, Hart T, et al: Effect of citicoline on functional and cognitive status among patients with traumatic brain injury: Citicoline Brain Injury Treatment Trial (COBRIT). JAMA. 308:1993–2000. 2012.PubMed/NCBI View Article : Google Scholar

83 

Narayan RK, Maas AI, Marshall LF, Servadei F, Skolnick BE and Tillinger MN: rFVIIa Traumatic ICH Study Group. Recombinant factor VIIA in traumatic intracerebral hemorrhage: Results of a dose-escalation clinical trial. Neurosurgery. 62:776–786; discussion 786-778. 2008.PubMed/NCBI View Article : Google Scholar

84 

Vulliamy P, Gillespie S, Gall LS, Green L, Brohi K and Davenport RA: Platelet transfusions reduce fibrinolysis but do not restore platelet function during trauma hemorrhage. J Trauma Acute Care Surg. 83:388–397. 2017.PubMed/NCBI View Article : Google Scholar

85 

Anglin CO, Spence JS, Warner MA, Paliotta C, Harper C, Moore C, Sarode R, Madden C and Diaz-Arrastia R: Effects of platelet and plasma transfusion on outcome in traumatic brain injury patients with moderate bleeding diatheses. J Neurosurg. 118:676–686. 2013.PubMed/NCBI View Article : Google Scholar

86 

Nair PM, Pidcoke HF, Cap AP and Ramasubramanian AK: Effect of cold storage on shear-induced platelet aggregation and clot strength. J Trauma Acute Care Surg. 77 (3 Suppl 2):S88–S93. 2014.PubMed/NCBI View Article : Google Scholar

87 

Etemadrezaie H, Baharvahdat H, Shariati Z, Lari SM, Shakeri MT and Ganjeifar B: The effect of fresh frozen plasma in severe closed head injury. Clin Neurol Neurosurg. 109:166–171. 2007.PubMed/NCBI View Article : Google Scholar

88 

Robertson CS, Hannay HJ, Yamal JM, Gopinath S, Goodman JC and Tilley BC: Epo Severe TBI Trial Investigators. Baldwin A, Rivera Lara L, Saucedo-Crespo H, et al: Effect of erythropoietin and transfusion threshold on neurological recovery after traumatic brain injury: A randomized clinical trial. JAMA. 312:36–47. 2014.PubMed/NCBI View Article : Google Scholar

89 

Lelubre C, Bouzat P, Crippa IA and Taccone FS: Anemia management after acute brain injury. Crit Care. 20(152)2016.PubMed/NCBI View Article : Google Scholar

90 

Fujiwara Y, Kitano H, Yamamoto T, Kokubun S and Hidai C: Activation peptide of coagulation factor IX improves the prognosis after traumatic brain injury. Biochem Biophys Res Commun. 569:35–40. 2021.PubMed/NCBI View Article : Google Scholar

91 

Kramer AH, Deis N, Ruddell S, Couillard P, Zygun DA, Doig CJ and Gallagher C: Decompressive craniectomy in patients with traumatic brain injury: Are the usual indications congruent with those evaluated in clinical trials? Neurocrit Care. 25:10–19. 2016.PubMed/NCBI View Article : Google Scholar

92 

Spahn DR, Bouillon B, Cerny V, Duranteau J, Filipescu D, Hunt BJ, Komadina R, Maegele M, Nardi G, Riddez L, et al: The European guideline on management of major bleeding and coagulopathy following trauma: Fifth edition. Crit Care. 23(98)2019.PubMed/NCBI View Article : Google Scholar

93 

Muradashvili N and Lominadze D: Role of fibrinogen in cerebrovascular dysfunction after traumatic brain injury. Brain Inj. 27:1508–1515. 2013.PubMed/NCBI View Article : Google Scholar

94 

Theusinger OM, Baulig W, Asmis LM, Seifert B and Spahn DR: In vitro factor XIII supplementation increases clot firmness in Rotation Thromboelastometry (ROTEM). Thromb Haemost. 104:385–391. 2010.PubMed/NCBI View Article : Google Scholar

95 

Gerlach R, Raabe A, Zimmermann M, Siegemund A and Seifert V: Factor XIII deficiency and postoperative hemorrhage after neurosurgical procedures. Surg Neurol. 54:260–264; discussion 264-265. 2000.PubMed/NCBI View Article : Google Scholar

96 

Steiner T, Poli S, Griebe M, Hüsing J, Hajda J, Freiberger A, Bendszus M, Bösel J, Christensen H, Dohmen C, et al: Fresh frozen plasma versus prothrombin complex concentrate in patients with intracranial haemorrhage related to vitamin K antagonists (INCH): A randomised trial. Lancet Neurol. 15:566–573. 2016.PubMed/NCBI View Article : Google Scholar

97 

Pollack CV Jr, Reilly PA, Eikelboom J, Glund S, Verhamme P, Bernstein RA, Dubiel R, Huisman MV, Hylek EM, Kamphuisen PW, et al: Idarucizumab for dabigatran reversal. N Engl J Med. 373:511–520. 2015.PubMed/NCBI View Article : Google Scholar

98 

Connolly SJ, Crowther M, Eikelboom JW, Gibson CM, Curnutte JT, Lawrence JH, Yue P, Bronson MD, Lu G, Conley PB, et al: Full study report of andexanet alfa for bleeding associated with factor Xa Inhibitors. N Engl J Med. 380:1326–1335. 2019.PubMed/NCBI View Article : Google Scholar

99 

Frontera JA, Lewin JJ III, Rabinstein AA, Aisiku IP, Alexandrov AW, Cook AM, Del Zoppo GJ, Kumar M, Peerschke EI, Stiefel MF, et al: Guideline for reversal of antithrombotics in intracranial hemorrhage: Executive summary. A statement for healthcare professionals from the neurocritical care society and the society of critical care medicine. Crit Care Med. 44:2251–2257. 2016.PubMed/NCBI View Article : Google Scholar

100 

Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz MA, Fini ME and Lo EH: Effects of matrix metalloproteinase-9 gene knock-out on the proteolysis of blood-brain barrier and white matter components after cerebral ischemia. J Neurosci. 21:7724–7732. 2001.PubMed/NCBI View Article : Google Scholar

101 

Zhao BQ, Wang S, Kim HY, Storrie H, Rosen BR, Mooney DJ, Wang X and Lo EH: Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med. 12:441–445. 2006.PubMed/NCBI View Article : Google Scholar

102 

Shukla M, Sekhon UD, Betapudi V, Li W, Hickman DA, Pawlowski CL, Dyer MR, Neal MD, McCrae KR and Sen Gupta A: In vitro characterization of SynthoPlate™ (synthetic platelet) technology and its in vivo evaluation in severely thrombocytopenic mice. J Thromb Haemost. 15:375–387. 2017.PubMed/NCBI View Article : Google Scholar

103 

Fitzpatrick GM, Cliff R and Tandon N: Thrombosomes: A platelet-derived hemostatic agent for control of noncompressible hemorrhage. Transfusion. 53 (Suppl 1):100S–106S. 2013.PubMed/NCBI View Article : Google Scholar

104 

Ahmed S, Venigalla H, Mekala HM, Dar S, Hassan M and Ayub S: Traumatic brain injury and neuropsychiatric complications. Indian J Psychol Med. 39:114–121. 2017.PubMed/NCBI View Article : Google Scholar

105 

Chen H, Yuan F, Chen SW, Guo Y, Wang G, Deng ZF and Tian HL: Predicting posttraumatic hydrocephalus: Derivation and validation of a risk scoring system based on clinical characteristics. Metab Brain Dis. 32:1427–1435. 2017.PubMed/NCBI View Article : Google Scholar

106 

Wang XC, Gao SJ, Zhuo SL, Weng CL, Feng HW, Lin J, Lin XS and Huang L: Predictive factors for cerebrocardiac syndrome in patients with severe traumatic brain injury: A retrospective cohort study. Front Neurol. 4(1192756)2023.PubMed/NCBI View Article : Google Scholar

107 

Campwala I, Guyette FX, Brown JB, Yazer MH, Daley BJ, Miller RS, Harbrecht BG, Claridge JA, Phelan HA, Eastridge B, et al: Evaluation of critical care burden following traumatic injury from two randomized controlled trials. Sci Rep. 13(1106)2023.PubMed/NCBI View Article : Google Scholar

108 

Nicolai L and Massberg S: Platelets as key players in inflammation and infection. Curr Opin Hematol. 27:34–40. 2020.PubMed/NCBI View Article : Google Scholar

109 

Nicolai L, Leunig A, Brambs S, Kaiser R, Weinberger T, Weigand M, Muenchhoff M, Hellmuth JC, Ledderose S, Schulz H, et al: Immunothrombotic Dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation. 142:1176–1189. 2020.PubMed/NCBI View Article : Google Scholar

110 

Passacquale G, Vamadevan P, Pereira L, Hamid C, Corrigall V and Ferro A: Monocyte-platelet interaction induces a pro-inflammatory phenotype in circulating monocytes. PLoS One. 6(e25595)2011.PubMed/NCBI View Article : Google Scholar

111 

Bozza FA, Shah AM, Weyrich AS and Zimmerman GA: Amicus or adversary: Platelets in lung biology, acute injury, and inflammation. Am J Respir Cell Mol Biol. 40:123–134. 2009.PubMed/NCBI View Article : Google Scholar

112 

Cole E, Davenport R, Willett K and Brohi K: Tranexamic acid use in severely injured civilian patients and the effects on outcomes: A prospective cohort study. Ann Surg. 261:390–394. 2015.PubMed/NCBI View Article : Google Scholar

113 

Einersen PM, Moore EE, Chapman MP, Moore HB, Gonzalez E, Silliman CC, Banerjee A and Sauaia A: Rapid thrombelastography thresholds for goal-directed resuscitation of patients at risk for massive transfusion. J Trauma Acute Care Surg. 82:114–119. 2017.PubMed/NCBI View Article : Google Scholar

114 

Ekeh AP, Dominguez KM, Markert RJ and McCarthy MC: Incidence and risk factors for deep venous thrombosis after moderate and severe brain injury. J Trauma. 68:912–915. 2010.PubMed/NCBI View Article : Google Scholar

115 

Ivanov II, Apta BHR, Bonna AM and Harper MT: Platelet P-selectin triggers rapid surface exposure of tissue factor in monocytes. Sci Rep. 9(13397)2019.PubMed/NCBI View Article : Google Scholar

116 

Teichman AL, Cotton BA, Byrne J, Dhillon NK, Berndtson AE, Price MA, Johns TJ, Ley EJ, Costantini T and Haut ER: Approaches for optimizing venous thromboembolism prevention in injured patients: Findings from the consensus conference to implement optimal venous thromboembolism prophylaxis in trauma. J Trauma Acute Care Surg. 94:469–478. 2023.PubMed/NCBI View Article : Google Scholar

117 

Knudson MM, Moore EE, Kornblith LZ, Shui AM, Brakenridge S, Bruns BR, Cipolle MD, Costantini TW, Crookes BA, Haut ER, et al: Challenging traditional paradigms in posttraumatic pulmonary thromboembolism. JAMA Surg. 157(e216356)2022.PubMed/NCBI View Article : Google Scholar

118 

Stein SC, Chen XH, Sinson GP and Smith DH: Intravascular coagulation: A major secondary insult in nonfatal traumatic brain injury. J Neurosurg. 97:1373–1377. 2002.PubMed/NCBI View Article : Google Scholar

119 

Chelladurai Y, Stevens KA, Haut ER, Brotman DJ, Sharma R, Shermock KM, Kebede S, Singh S and Segal JB: Venous thromboembolism prophylaxis in patients with traumatic brain injury: A systematic review. F1000Res. 2(132)2013.PubMed/NCBI View Article : Google Scholar

120 

Verhamme P, Yi BA, Segers A, Salter J, Bloomfield D, Büller HR, Raskob GE and Weitz JI: ANT-005 TKA Investigators. Abelacimab for prevention of venous thromboembolism. N Engl J Med. 385:609–617. 2021.PubMed/NCBI View Article : Google Scholar

121 

Dübendorfer C, Billeter AT, Seifert B, Keel M and Turina M: Serial lactate and admission SOFA scores in trauma: An analysis of predictive value in 724 patients with and without traumatic brain injury. Eur J Trauma Emerg Surg. 39:25–34. 2013.PubMed/NCBI View Article : Google Scholar

122 

Raj R, Siironen J, Kivisaari R, Hernesniemi J and Skrifvars MB: Predicting outcome after traumatic brain injury: Development of prognostic scores based on the IMPACT and the APACHE II. J Neurotrauma. 31:1721–1732. 2014.PubMed/NCBI View Article : Google Scholar

123 

MacLeod JB, Lynn M, McKenney MG, Cohn SM and Murtha M: Early coagulopathy predicts mortality in trauma. J Trauma. 55:39–44. 2003.PubMed/NCBI View Article : Google Scholar

124 

McCully SP, Fabricant LJ, Kunio NR, Groat TL, Watson KM, Differding JA, Deloughery TG and Schreiber MA: The International Normalized Ratio overestimates coagulopathy in stable trauma and surgical patients. J Trauma Acute Care Surg. 75:947–953. 2013.PubMed/NCBI View Article : Google Scholar

125 

Mann KG, Butenas S and Brummel K: The dynamics of thrombin formation. Arterioscler Thromb Vasc Biol. 23:17–25. 2003.PubMed/NCBI View Article : Google Scholar

126 

Haas T, Fries D, Tanaka KA, Asmis L, Curry NS and Schöchl H: Usefulness of standard plasma coagulation tests in the management of perioperative coagulopathic bleeding: Is there any evidence? Br J Anaesth. 114:217–224. 2015.PubMed/NCBI View Article : Google Scholar

127 

Schöchl H, Solomon C, Traintinger S, Nienaber U, Tacacs-Tolnai A, Windhofer C, Bahrami S and Voelckel W: Thromboelastometric (ROTEM) findings in patients suffering from isolated severe traumatic brain injury. J Neurotrauma. 28:2033–2041. 2011.PubMed/NCBI View Article : Google Scholar

128 

Inaba K, Rizoli S, Veigas PV, Callum J, Davenport R, Hess J and Maegele M: Viscoelastic Testing in Trauma Consensus Panel. 2014 Consensus conference on viscoelastic test-based transfusion guidelines for early trauma resuscitation: Report of the panel. J Trauma Acute Care Surg. 78:1220–1229. 2015.PubMed/NCBI View Article : Google Scholar

129 

Connelly CR, Van PY, Hart KD, Louis SG, Fair KA, Erickson AS, Rick EA, Simeon EC, Bulger EM, Arbabi S, et al: Thrombelastography-Based dosing of enoxaparin for thromboprophylaxis in trauma and surgical patients: A Randomized clinical trial. JAMA Surg. 151(e162069)2016.PubMed/NCBI View Article : Google Scholar

130 

Chang R, Cardenas JC, Wade CE and Holcomb JB: Advances in the understanding of trauma-induced coagulopathy. Blood. 128:1043–1049. 2016.PubMed/NCBI View Article : Google Scholar

131 

Wikkelsø A, Wetterslev J, Møller AM and Afshari A: Thromboelastography (TEG) or thromboelastometry (ROTEM) to monitor haemostatic treatment versus usual care in adults or children with bleeding. Cochrane Database Syst Rev. 2016(Cd007871)2016.PubMed/NCBI View Article : Google Scholar

132 

Rossaint R, Afshari A, Bouillon B, Cerny V, Cimpoesu D, Curry N, Duranteau J, Filipescu D, Grottke O, Grønlykke L, et al: The European guideline on management of major bleeding and coagulopathy following trauma: Sixth edition. Crit Care. 27(80)2023.PubMed/NCBI View Article : Google Scholar

133 

Woolley T, Gwyther R, Parmar K, Kirkman E, Watts S, Midwinter M, Lucca JD and Hunt BJ: A prospective observational study of acute traumatic coagulopathy in traumatic bleeding from the battlefield. Transfusion. 60 (Suppl 3):S52–S61. 2020.PubMed/NCBI View Article : Google Scholar

134 

Dunbar NM and Chandler WL: Thrombin generation in trauma patients. Transfusion. 49:2652–2660. 2009.PubMed/NCBI View Article : Google Scholar

135 

Davenport R, Manson J, De'Ath H, Platton S, Coates A, Allard S, Hart D, Pearse R, Pasi KJ, MacCallum P, et al: Functional definition and characterization of acute traumatic coagulopathy. Crit Care Med. 39:2652–2658. 2011.PubMed/NCBI View Article : Google Scholar

136 

Steiner LA, Czosnyka M, Piechnik SK, Smielewski P, Chatfield D, Menon DK and Pickard JD: Continuous monitoring of cerebrovascular pressure reactivity allows determination of optimal cerebral perfusion pressure in patients with traumatic brain injury. Crit Care Med. 30:733–738. 2002.PubMed/NCBI View Article : Google Scholar

137 

Depreitere B, Güiza F, Van den Berghe G, Schuhmann MU, Maier G, Piper I and Meyfroidt G: Pressure autoregulation monitoring and cerebral perfusion pressure target recommendation in patients with severe traumatic brain injury based on minute-by-minute monitoring data. J Neurosurg. 120:1451–1457. 2014.PubMed/NCBI View Article : Google Scholar

138 

Chesnut RM, Marshall LF, Klauber MR, Blunt BA, Baldwin N, Eisenberg HM, Jane JA, Marmarou A and Foulkes MA: The role of secondary brain injury in determining outcome from severe head injury. J Trauma. 34:216–222. 1993.PubMed/NCBI View Article : Google Scholar

139 

Bernard F, Barsan W, Diaz-Arrastia R, Merck LH, Yeatts S and Shutter LA: Brain Oxygen Optimization in Severe Traumatic Brain Injury (BOOST-3): A multicentre, randomised, blinded-endpoint, comparative effectiveness study of brain tissue oxygen and intracranial pressure monitoring versus intracranial pressure alone. BMJ Open. 12(e060188)2022.PubMed/NCBI View Article : Google Scholar

140 

Okonkwo DO, Shutter LA, Moore C, Temkin NR, Puccio AM, Madden CJ, Andaluz N, Chesnut RM, Bullock MR, Grant GA, et al: Brain oxygen optimization in severe traumatic brain injury phase-II: A phase II Randomized trial. Crit Care Med. 45:1907–1914. 2017.PubMed/NCBI View Article : Google Scholar

141 

Zeiler FA, Thelin EP, Helmy A, Czosnyka M, Hutchinson PJA and Menon DK: A systematic review of cerebral microdialysis and outcomes in TBI: Relationships to patient functional outcome, neurophysiologic measures, and tissue outcome. Acta Neurochir (Wien). 159:2245–2273. 2017.PubMed/NCBI View Article : Google Scholar

142 

Robinson MB, Shin P, Alunday R, Cole C, Torbey MT and Carlson AP: Decision-making for decompressive craniectomy in traumatic brain injury aided by multimodality monitoring: Illustrative case. J Neurosurg Case Lessons. 1(CASE2197)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2024
Volume 21 Issue 5

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hou H, Qu Z, Liu R, Jiang B, Wang L and Li A: Traumatic brain injury: Advances in coagulopathy (Review). Biomed Rep 21: 156, 2024.
APA
Hou, H., Qu, Z., Liu, R., Jiang, B., Wang, L., & Li, A. (2024). Traumatic brain injury: Advances in coagulopathy (Review). Biomedical Reports, 21, 156. https://doi.org/10.3892/br.2024.1844
MLA
Hou, H., Qu, Z., Liu, R., Jiang, B., Wang, L., Li, A."Traumatic brain injury: Advances in coagulopathy (Review)". Biomedical Reports 21.5 (2024): 156.
Chicago
Hou, H., Qu, Z., Liu, R., Jiang, B., Wang, L., Li, A."Traumatic brain injury: Advances in coagulopathy (Review)". Biomedical Reports 21, no. 5 (2024): 156. https://doi.org/10.3892/br.2024.1844