Next Article in Journal
Medicines—Aims and Scope Updates
Previous Article in Journal
Human Leucocyte Antigen Genetics in Idiosyncratic Drug-Induced Liver Injury with Evidence Based on the Roussel Uclaf Causality Assessment Method
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Modern Approach to the Treatment of Traumatic Brain Injury

1
Department of Hospital Surgery, Anesthesiology and Reanimatology, Semey Medical University, Semey 071400, Kazakhstan
2
Department of General Practitioners, Semey Medical University, Semey 071400, Kazakhstan
3
Department of Pediatric Surgery, Semey Medical University, Semey 071400, Kazakhstan
4
Committee for Medical and Pharmaceutical Control of the Ministry of Health of the Republic of Kazakhstan for East Kazakhstan Region, Ust-Kamenogorsk 070004, Kazakhstan
5
Pavlodar Branch of Semey Medical University, Pavlodar S03Y3M1, Kazakhstan
6
Department Psychiatry and Narcology, Semey Medical University, Semey 071400, Kazakhstan
7
Department of Therapy, Astana Medical University, Astana 010000, Kazakhstan
8
Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978, Israel
*
Author to whom correspondence should be addressed.
Medicines 2024, 11(5), 10; https://doi.org/10.3390/medicines11050010
Submission received: 5 March 2024 / Revised: 18 April 2024 / Accepted: 27 April 2024 / Published: 30 April 2024
(This article belongs to the Section Neurology and Neurologic Diseases)

Abstract

:
Background: Traumatic brain injury manifests itself in various forms, ranging from mild impairment of consciousness to severe coma and death. Traumatic brain injury remains one of the leading causes of morbidity and mortality. Currently, there is no therapy to reverse the effects associated with traumatic brain injury. New neuroprotective treatments for severe traumatic brain injury have not achieved significant clinical success. Methods: A literature review was performed to summarize the recent interdisciplinary findings on management of traumatic brain injury from both clinical and experimental perspective. Results: In the present review, we discuss the concepts of traditional and new approaches to treatment of traumatic brain injury. The recent development of different drug delivery approaches to the central nervous system is also discussed. Conclusions: The management of traumatic brain injury could be aimed either at the pathological mechanisms initiating the secondary brain injury or alleviating the symptoms accompanying the injury. In many cases, however, the treatment should be complex and include a variety of medical interventions and combination therapy.

1. Introduction

The approach to brain injury has evolved with advances in medical science and the application of evidence-based medicine. Over the past 20 years, more than 30 traumatic brain injury (TBI) guidelines have been developed and updated by various organizations [1,2]. However, there is currently no ultimate and effective guideline or treatment protocol for TBI. Thus, the positive effects observed in randomized clinical trials (RCTs) of various pharmacological treatments of severe TBI were small, typically with <10% difference between groups [3,4].
Effective treatment requires the identification of key therapeutic targets. There are two general approaches to the development of TBI management today. The first, the “traditional” approach is mainly based on neuroprotection and utilizes an identification and targeting of the key mechanisms involved in the development of secondary injury, whether it be severe or mild TBI. In this traditional approach, treatment is usually initiated as soon as possible after the injury since delaying neuroprotective therapy is thought to reduce its effectiveness. Traditional therapy often targets key initiators of the secondary injury cascade. The second, an “alternative” approach being explored in clinical trials for patients with mild TBI is symptomatic rather than mechanistic treatment aimed at symptoms such as headache, sleep disturbances, vestibular/oculomotor disturbances, post-traumatic stress disorder, cognitive dysfunction, or other secondary consequences [5].
This review discusses several current research concepts and strategies for TBI management, both “traditional” and “alternative”. The general overview of the discussed strategies and methods of TBI treatment is outlined in Table 1 below.

2. Concepts and Strategies for TBI Therapy

Symptomatic treatment is often used in some emergency cases of severe TBI. Based on clinical or physiological manifestations, particular forms of therapy (such as hyperosmolar agents, barbiturates, hypothermia, and craniectomy) are used to alleviate intracranial hypertension [6,7,8]. However, in finding an effective treatment for severe TBI the main research attention has always been focused on novel neuroprotective agents [9,10]. Thus, using a traditional, “mechanistic” approach, Kochanek et al. [9], Mondello et al. [11], and Yang et al. [12] have attempted to develop new neuroprotective therapies for TBI treatment. Operation brain trauma therapy (OBTT) is just one approach to establishing a TBI screening consortium. The OBTT uses a scoring matrix that includes 22 points per animal model assessed on several parameters including motor, cognitive, histological, and serum biomarkers [11]. Using the OBTT, a total of 12 drugs have been tested, namely nicotinamide, erythropoietin, cyclosporin A, simvastatin, levetiracetam, glibenclamide, Kollidon@VA-64, amantadine, minocycline, axiostatin (E64d), and P7C3-A20. Two drugs, levetiracetam and glibenclamide, appeared to be the most promising. Thus, levetiracetam improved multiple outcomes in two experimental models of TBI, namely fluid percussion brain injury (FPI) and controlled cortical impact (CCI) [13], while glibenclamide displayed relatively stable benefit in CCI [14,15]; however, the above benefits were largely limited to the particular experimental model.
The goal of novel approaches to TBI management is to identify and develop robust therapy in a pre-clinical study, determine the optimal dosage and treatment regimens, and implement the most effective methods in medical practice. Therefore, optimal communication between pre-clinical and clinical studies appears to be essential for the successful development and implementation of new methods of TBI management and improved patient outcomes.

3. Secondary Brain Injury in TBI

Despite significant improvements in resuscitation after TBI, one of the inevitable consequences of neurotrauma is secondary brain injury. Direct mechanical impact can cause force dynamics to be transmitted through the dura mater, cerebrospinal fluid, and brain parenchyma. This can cause axons to become dislocated, cell membranes to break down, and fluid to shift, leading to swelling of the brain tissue. This primary trauma eventually develops into a secondary trauma response [16]. Although currently there is no FDA-approved drug to prevent or treat damage secondary to neurotrauma [17], understanding the mechanisms of pathophysiology of secondary brain injury is important for the successful treatment of traumatic brain injury.
Biochemical, cellular, and physiological events occurring during primary injury often develop into delayed and prolonged secondary injuries that can last from hours to years. Secondary brain injury can cause many harmful effects such as cerebral edema, blood–brain barrier (BBB) disruption, increased intracranial pressure, metabolic dysfunctions, excitotoxicity, oxidative and cellular apoptosis, or necrosis, ultimately leading to neurological dysfunctions [18].
A cascade of secondary damage typically unfolds over time, ranging from minutes to days after the initial damage. It may be influenced by many factors such as systemic hypotension, hypoxemia, increased intracranial pressure, and metabolic imbalance [10,19]. In addition, the severity of the secondary injury and its impact on disease outcome depends on various factors, including the nature and extent of the primary injury, as well as the effectiveness of medical interventions aimed at mitigating the progression of the damage [18,20,21,22,23,24].
Secondary damage from neurotrauma is associated with endoplasmic reticulum (ER) and mitochondrial dysfunction, as well as oxidative stress [10,25,26,27,28]. Stress on these organelles triggers a cascade of events that ultimately activate neuroinflammatory pathways [28,29]. Signs of neuroinflammation or CNS immune response begin to develop soon after the initial injury. Some obvious changes include ventricular enlargement, edema, and white and/or gray matter atrophy, while microscopic apoptosis, autophagy, axonal damage, and necrosis are visualized [30,31].

4. Emergency Care for TBI

4.1. Head Position

Lifting the head of a patient with a traumatic brain injury usually has quick consequences. The intracranial pressure (ICP) is reduced due to the displacement of cerebrospinal fluid (CSF) from the intracranial space and increasing of venous outflow [32]. Although mean carotid pressure decreases with head elevation, ICP decreases while cerebral blood flow (CBF) does not change [33].

4.2. Airway Management

4.2.1. Tracheostomy

Tracheostomy is often performed on TBI patients, mainly because of many associated beneficial effects, such as protection of airways from aspiration, enhanced comfort of the patient, decreased requirement for sedation, improved pulmonary hygiene, and more effective secretion suction [34]. Tracheostomy decreases the duration of stay in the intensive care unit, as well as the number of days on an artificial ventilation machine [34].
There is ongoing debate about the benefits of early tracheostomy over late tracheostomy [35,36]. Early tracheostomy could be associated with more ventilator-free days [36,37]. Thus, several recent studies demonstrated that early tracheostomy significantly decreases both pulmonary morbidity and critical care resource utilization [37,38,39,40,41,42].

4.2.2. Hyperventilation

Hyperventilation reduces ICP by lowering the arterial partial pressure of carbon dioxide (PaCO2), which subsequently leads to vasoconstriction. This sequence of events ultimately leads to a decrease in brain blood volume [43]. Prophylactic hyperventilation is generally not recommended since vasoconstriction reduces the CBF. As a result of vasoconstriction, focal foci of ischemia may occur, especially in areas of preserved autoregulation [44].

4.2.3. Ventilator-Associated Pneumonia

Early onset ventilator-associated pneumonia (EOVAP) is considered a common complication in TBI patients, with a frequency of occurrence of 24–60% [45,46]. EOVAP could impair cerebral oxygenation and its occurrence is a factor in worsening neurological outcomes [46]. The risk factors of EOVAP in TBI patients are not fully determined, and the effects of various pre-hospital methods of airway management techniques on EOVAP occurrence in this population have not yet been investigated. However, there is some evidence that pre-hospital airway management does not significantly affect the occurrence of EOVAP in patients with severe TBI [47]. Similarly, despite the increased duration of mechanical ventilation and prolonged stay in the intensive care unit, pre-hospital airway management appears to have no significant effect on mortality or long-term neurological outcomes [47].

4.3. Prevention of Seizures

Current recommendations for TBI treatment state that prophylactic administration of antiepileptic drugs for one week could be used to prevent early seizures. However, to date, there has been no proven benefit in preventing late seizures after TBI, and therefore antiepileptic therapy is usually discontinued after 7 days [48].
A large prospective randomized controlled study found no difference in the incidence of early post-traumatic seizures between a group of patients treated with phenytoin and another group treated with levetiracetam [49].

4.4. Sedation and Induced Coma

One of the last steps of maximal treatment is to put a patient into a drug-induced coma, usually by infusion of benzodiazepines such as midazolam or infusion of barbiturates such as pentobarbital. Benzodiazepines and barbiturates exert their action by a significant reduction of the metabolic demands in the brain. Prophylactic use of barbiturates for seizure suppression is usually not recommended. However, the administration of barbiturates could be used in severe refractory intracranial hypertension after exhaustion of maximum medical and surgical therapy targeting reduction of intracranial pressure [43].
Reducing stress and adrenocortical response is also considered an important component of TBI treatment [50]. By reducing cerebral metabolism and oxygen consumption in a dose-dependent manner [51], sedatives can reduce metabolic stress in acutely injured brain tissue and lead to a decrease in the ICP. Achieving an adequate level of sedation is of paramount importance, since it minimizes the length of hospital stay, the number of days on a ventilator, and the frequency of delirium and promotes early mobilization [52]. The Brain Trauma Foundation (BTF) recommendations [6] regarding the use of sedatives and analgesics are as follows:
  • Administration of barbiturates to suppress seizures and as a prophylaxis for intracranial hypertension is not recommended (grade IIB).
  • High doses of barbiturates are recommended to control intracranial pressure refractory to maximum standard surgical and medical treatments while maintaining hemodynamic stability.
  • Although propofol can be used to control intracranial pressure, it is not recommended for reducing mortality or six-month outcomes.

4.5. Hypothermia

Oxidative stress is believed to be a secondary effect of TBI. Therapeutic hypothermia in infants and children has been shown to reduce oxidative damage [53].
There are several plausible mechanisms by which hypothermia may mitigate the effects of TBI, including a decrease in intracranial pressure, a decrease in the innate inflammatory response, and a decrease in the cerebral metabolic rate [49,54].
The multicenter, non-blinded RCT Eurotherm3235 (2015) is the largest study of hypothermia in patients with intracranial hypertension (>20 mmHg) after TBI. This study has demonstrated that therapeutic mild hypothermia [48,49,50,51] (32–35 °C) combined with standard therapy to reduce ICP resulted in a slightly increased mortality rate and poor functional outcomes compared with patients receiving standard therapy alone. In addition, an unfavorable association between hypothermia and the progression of multiple organ failure has been observed [55,56].
It should be stressed that deep hypothermia (below 30 °C) does not appear to have a beneficial effect on TBI, while mild to moderate hypothermia (32–35 °C) appears to be neuroprotective [57,58].
Hypothermia (32–34 °C) reduces cerebral metabolic rate, preserves tissues in the face of metabolic problems, and reduces intracranial pressure. There is insufficient evidence to support prophylactic hypothermia; although not currently a standard therapy, it may be an option for a multimodal approach, as supported by TBI guidelines [59]. The use of hypothermia is not a standard recommendation in the management of patients with TBI [6]. Patients with TBI treated with hypothermia show little improvement in outcomes. However, in patients with severe TBI, fever should be actively controlled [60].

4.6. Blood Pressure and Cerebral Perfusion Pressure (CPP)

For optimal perfusion of organs and tissues, the systolic blood pressure should be maintained at a range of 110–130 mm Hg while diastolic blood pressure should not be allowed to decrease below 60 mm Hg. Increased mortality (about 35%) of TBI patients with systolic blood pressure (SBP) ≤ 85 mm Hg has been reported, compared to only 6% mortality in patients with higher SBP [61].
For resuscitation, intravenous administration of fluids and vasopressors should be used [62]. The ideal intravenous fluid for TBI patients remains a matter of debate (please see “Fluid management” below). However, hypotonic fluids should be avoided to prevent volume overload and cerebral edema [63,64].
The 4th edition of the BTF manual recommends [4]:
  • To reduce mortality and improve the outcomes (level III), maintain SBP at ≥100 mm Hg for patients aged 50 to 69 years or at a level ≥110 mm Hg or higher for patients aged 15 to 49 or older than 70.
  • The recommended CPP target for survival and good outcomes is 60 to 70 mm Hg. The optimal CPP may depend on the patient’s autoregulatory status (level IIB).
  • Aggressive attempts to maintain CPP above 70 mm Hg with fluids and vasopressors should be avoided.

4.7. Fluid Management

Available evidence suggests that the volume of fluid administered, rather than the choice of the fluid itself, plays an important role in outcomes after TBI [54,63]. Saline is the most common crystalloid solution used in TBI patients, but Ringer’s lactate could be used as an alternative. Crystalloid solutions generally do not expand well, and about 70–80% of their infused volume usually reaches the interstitial space within 20 min of infusion, contributing to general systemic tissue edema. During the breaching of the blood–brain barrier (BBB), the significant passive spread of the crystalloid solution to the interstitium of the brain can occur and this, in turn, can exacerbate the cerebral edema and increase the intracranial pressure, especially when using hypotonic solutions. In addition, the infusion of large volumes of saline can lead to unfavorable hyperchloremic metabolic acidosis, which is dangerous in TBI. Balanced crystalloid solutions can be a better alternative to saline [65].
Hypertonic crystalloid solutions, such as hypertonic saline, glycerol, urea, sorbitol, and mannitol are often used to reduce brain swelling in TBI. The hypertonic solutions decrease intracranial pressure and promote the shrinkage of the brain tissue by establishing a strong trans-epithelial osmotic gradient and directing the water flow from the brain tissue to the intravascular compartment. However, some of the above solutions have significant side effects. For example, the use of urea could be associated with nausea, vomiting, diarrhea, hemoglobinuria, and rebound intracranial hypertension [66]. Sorbitol and glycerol are often associated with a significant increase in blood glucose levels which could be additionally harmful to TBI patients [66]. Hypertonic saline has been widely used for the treatment of TBI since 1919 [67]. The solution is available in different concentrations (3.0–23.4%); however, there is no conclusion on the effectiveness and safety of particular concentrations in TBI treatment. Hypertonic saline in the 3.0–7.5% range could be infused without dilution or in combination with isotonic crystalloids or colloids for hypovolemic shock resuscitation [68]. Mannitol appears to be effective in reversing acute brain swelling; however, its effectiveness in the ongoing management of TBI remains unclear [69]. The use of mannitol has been associated with hypotension [66,70,71], rebound increase in intracranial pressure [72], and renal toxicity [73]. Overall, mannitol treatment was reported to be associated with a detrimental effect on mortality when compared to hypertonic saline [69].
Colloid solutions do not appear to provide additional benefits, and the Saline versus Albumin Fluid Evaluation SAFE) study found increased mortality in patients treated with albumin compared with saline [74]. In many patients (9–23%), TBI is linked to acute renal injury and frequently results in increased mortality [75]. Additionally, it has been discovered that administration of colloids raises the risk of acute kidney injury and increases the need for renal replacement treatment in severe TBI. According to a recent assessment, colloids are not more effective than crystalloids in terms of overall mortality, particularly when it comes to patients who have had surgery, trauma, or burns [76]. However, colloid administration in patients with preserved renal function may be considered [76].

4.8. Tranexamic Acid

Administration of a hemostatic agent, tranexamic acid, is necessary for the prevention of hemorrhages in TBI. Currently, available data from the CRASH-3 study suggest that the administration of tranexamic acid to patients with mild or moderate TBI resulted in a reduction of mortality by 20% [77]. Also, the meta-analysis of Yokobori et al. [78] suggested that treatment with tranexamic acid displayed a trend toward reduced mortality from head injuries in patients with severe TBI without a significant incidence of thromboembolic complications. However, Bossers et al. [79] demonstrated that the pre-hospital administration of tranexamic acid was associated with a higher mortality rate in patients with severe TBI, especially isolated TBI.
The meta-analysis of Huang et al. [80] demonstrated that administration of tranexamic acid during the initial 3 h after the trauma may reduce mean hemorrhage volume. However, the reduction of mean hemorrhage volume was not observed when the drug was administered more than 3 h after trauma [80]. Therefore, the timing of tranexamic acid administration appears to be one of the crucial factors affecting its hemostatic effect.
Thus, the results of current studies do not provide a clear answer to the question of whether the use of tranexamic acid reduces mortality and morbidity rates, especially in patients with severe TBI. However, treatment with tranexamic acid could be effective in patients with mild or moderate TBI.

5. Surgical Interventions for TBI

Decompressive craniectomy (DC) is an effective, lifesaving option for lowering intracranial pressure in TBI patients [81]. Most DCs are performed via a standard trauma flap shaped like a reverse question mark (RQM) [81]. As an alternative to an RQM, the safer and more effective Ludwig Kempe hemispherectomy incision (Kempe) could be used [82]. It has been shown that DC can be lifesaving for patients with TBI with a decrease in mortality [82]. The BTF guidelines recommend a larger frontotemporoparietal decompressive craniectomy, rather than a smaller one, to reduce mortality and improve neurological outcomes [6].
One of the consequences of DC is the need for cranioplasty (replacement of a bone defect with an artificial plate). Currently, there is no consensus on the timing of cranioplasty, cranioplasty material (e.g., titanium, polyethylene, methyl methacrylate, and hydroxyapatite), or the use of autologous bone [83].
Cisternostomy is a novel surgical strategy for severe TBI management. Several reports demonstrated promising results with the use of cisternostomy compared to the traditional methods of TBI treatment [84,85,86].

6. Pharmacological Therapy of TBI

6.1. Corticosteroids

Corticosteroids reduce the production of CSF and exert antiedematous and anti-inflammatory effects in TBI. The first large-scale pharmacological trial of TBI was the randomization trial of corticosteroids after a major head injury (MRC corticosteroid randomization after significant head injury (CRASH), 2004). The patients with moderate to severe TBI were given a 48-h infusion with high doses of corticosteroids (methylprednisolone) or a placebo. In this multicenter study, 10,008 patients from 49 countries participated. The primary outcome of the study was 2-week mortality, which was higher in the treatment group (21.1%) than in the placebo group (17.9%). After 6 months of follow-up, there were 173 additional deaths in the treatment group (1248 vs. 1075) [87]. These results show there is no reduction in mortality with corticosteroid administration in the 2 weeks after head injury.

6.2. Progesterone

Following the failure of corticosteroids in TBI, pre-clinical animal studies have focused on the early administration of progesterone, a potent neurosteroid synthesized in the central nervous system. It has been demonstrated that the administration of progesterone reduces neuronal loss, cerebral edema, and behavioral disturbances after experimental TBI. However, large double-blind, placebo-controlled, multicenter phase III RCTs (SYNAPSE and PROTECT III) failed to demonstrate a significant positive effect of progesterone on patient mortality and functional outcomes, which led to the end of the enthusiasm generated by two previous single-center clinical trials [4,88,89].

6.3. Erythropoietin

Erythropoietin (EPO) is a glycoprotein that regulates hematopoiesis in the bone marrow, which is naturally produced in the kidneys after hypoxic stimulation. Animal studies have demonstrated that EPO can neutralize neuronal apoptosis, reduce the inflammatory response, and act as a neurotrophic factor, thereby alleviating the consequences of secondary brain damage in TBI [90].
A meta-analysis of six RCTs involving 1041 patients which examined the results of EPO treatment after acute (moderate or severe) TBI demonstrated that EPO administration significantly reduced mortality but failed to reduce the incidence of adverse outcomes and did not improve the functional outcome [90]. There were no significant differences in complication rates, including deep vein thrombosis, between the EPO treatment and the control groups [90]. In general, the use of EPO in severe TBI appears to be controversial since some individual studies demonstrated benefits on functional outcomes while other studies failed to find any significant difference in outcomes [3,88,89,91].

6.4. Amantadine

Amantadine is a dopamine agonist used in Parkinson’s disease. Amantadine can be distributed to the frontal lobes and acts as an N-methyl-D-aspartate (NMDA) receptor antagonist. It has been suggested that the administration of amantadine increases the external dopamine levels in the striatum and results in antagonism of NMDA receptor function [92], thereby protecting neurons from glutamate excitotoxicity in the acute phase of TBI. It has been shown that at doses of 200–400 mg/day, amantadine can improve arousal and cognition in TBI patients [93]. Other studies compared the effect of amantadine versus placebo; the results suggest that amantadine accelerated the pace of functional recovery in patients with severe TBI [94,95].

6.5. N-Acetylcysteine

It has been shown that in various animal models, N-acetylcysteine (NAC) has a significant neuroprotective effect by reducing the consequences of secondary neuronal damage. Rat models have confirmed the beneficial antioxidant properties of NAC in the treatment of brain injury [96]. It has been proposed that NAC exerts its neuroprotective effect through increasing levels of GSH, a combination of L-glutamic acid, L-cysteine, and glycine in the brain [97]. NAC administration maintains high levels of GSH in the brain, which acts as a free radical scavenger and as an antioxidant [96,97,98].
The effectiveness of NAC in treating patients with moderate blast-induced TBI was assessed in a double-blind, placebo-controlled human trial [99]. After receiving 2 g NAC twice a day for the first 4 days, the treatment group was given 1.5 g NAC twice a day for the next 3 days. After seven days of treatment, all patients were evaluated for headache, dizziness, memory loss, hearing loss, sleep difficulties, and neurocognitive impairment. Seven days after treatment, patients who received NAC within 24 h of the injury reported a significant improvement in these symptoms. In addition, in the treatment group, the chances for recovery were 86% [99].

6.6. Minocycline

It has been demonstrated that the tetracycline antibiotic minocycline (MINO), both alone and in combination with NAC, has neuroprotective qualities. The effectiveness of MINO in treating neurological problems resulting from traumatic brain injury has been demonstrated in rat models [100,101]. The memory and cognition of animals were enhanced in a mildly controlled cortical model by combined treatment with MINO + NAC, which also restored white matter by shielding oligodendrocytes [100].

6.7. Phenserine

In the treatment of TBI, the activity of phenserine reduces neuroinflammation, facilitates amyloid deposition, prevents apoptosis, and mitigates many different secondary injury mechanisms [102,103,104,105].

6.8. Calcium Channel Blockers

It has been shown that nimodipine, an L-type calcium channel blocker, improves outcomes for patients who have spontaneous subarachnoid hemorrhage [106]. A thorough study, however, found no statistically significant difference in death and morbidity rates between TBI patients treated with nimodipine and placebo [107].
Administration of an N-type calcium channel blocker ziconotide (SNX-111) between 15 min and 10 h after TBI has been shown to improve mitochondrial function in animals [108,109]. Another N-type calcium channel blocker SNX-185 has been reported to be neuroprotective when administered directly to hippocampal CA2 and CA3 24 h after TBI in rats [110]. More clinical research is required to determine the effectiveness and safety of calcium channel blockers in TBI treatment.

6.9. Antioxidants

The immunosuppressant cyclosporin A, a potent regulator of mitochondrial permeability transition pores (mPTPs), has been shown to have a neuroprotective effect in experimental TBI models [111]. However, a small randomized clinical trial of cyclosporin A in TBI unexpectedly demonstrated no improvement in neurological outcomes and biochemical parameters in patients with severe TBI [112].

6.10. Beta-Blockers

The benefits of early beta-blocker therapy in TBI have been demonstrated in several retrospective and prospective observational studies demonstrating a positive impact of beta-blockers on clinical outcomes and survival [113,114,115,116,117]. It has been hypothesized that the adrenergic storm caused by the primary stroke may exacerbate secondary brain injury through the mechanisms of cerebral vasoconstriction and subsequent ischemia [118,119]. Thus, hyperadrenergic activity is associated with an increased risk of death due to the exacerbation of secondary brain injury and induction of extracranial multiorgan dysfunction, especially cardiovascular, pulmonary, and inflammatory consequences [120,121].
Since the cerebral perfusion and subsequent oxygen delivery to the cerebral tissue are impaired due to the catecholamine-induced cerebral vasoconstriction [118,119], the use of beta-blockers to improve the brain environment in this context is based on physiological principles.
The early oral administration of beta-blocker propranolol in patients with isolated severe TBI results in improved survival and functional outcomes up to 6 months post-injury [122]. The above results provide support for the routine administration of beta-blockers as a part of standardized neuroresuscitation protocol. A meta-analysis by Ding et al. [123] demonstrated that the administration of beta-blockers after the TBI was safe and effective. Nevertheless, more studies are needed to investigate the effectiveness and safety of beta-blockers in TBI treatment.

6.11. Metformin

An antihyperglycemic agent metformin has been used clinically for decades as a treatment for diabetes. However, in addition to glucose regulation, metformin also stimulates neurogenesis [124] and has potent anti-inflammatory properties [125,126,127]. The above properties make metformin an interesting candidate for the treatment of CNS injuries. Indeed, several recent studies demonstrate the neuroprotective effects of metformin in various models of CNS injury [128,129,130].
Metformin significantly improves cognition after controlled cortical injury in mice, showing improved spatial learning and nest-building [131]. In addition, injured animals treated with metformin demonstrated increased ramification of microglial processes, indicating a reduction in neuroinflammation. Finally, in vitro, metformin treatment increased the activation of partitioning defective (Par1), a family of Ser/Thr kinases playing a key role in synaptic plasticity and neuroinflammation [131]. However, the improvement in behavioral outcomes of TBI by metformin treatment remains unclear, and the molecular mechanisms of metformin action are not well understood.

6.12. Cerebrolysin

The use of multimodal neuropeptide cerebrolysin in TBI treatment has been proposed by several studies [132,133,134,135,136]. It has been shown that cerebrolysin helps prevent secondary injury cascade by controlling oxidative stress, microglial activation, inflammation, and blood–brain barrier dysfunction [137]. However, the research on the effects of cerebrolysin in inoperable patients with severe TBI remains limited.
The recent meta-analysis demonstrated that intravenous administration of cerebrolysin significantly improved functional outcomes in patients with TBI measured by the Glasgow Outcome Scale and the modified Rankin Scale scores [138]. The cerebrolysin-treated group of inoperable patients with severe TBI demonstrated higher rates of recovery, as evidenced by an improvement in Glasgow Outcome Scale score and a shorter duration of stay in the hospital than the patients of the control group [139].

6.13. Vitamin D

Infectious complications, such as pneumonia and sepsis, are common in brain trauma patients and result in significant mortality in these patients. In addition, these patients have a high prevalence of vitamin D deficiency [140]. Since vitamin D deficiency is also associated with serious complications such as coma, slow neurological recovery, and persistent critical illness polyneuropathy in TBI patients [141,142,143], treatment with vitamin D could be of great importance in patients with TBI. Thus, Arabi et al. [144] suggest that vitamin D administration significantly reduces the mortality rate and inflammation in TBI patients.

7. Regenerative Treatment

7.1. Neurotrophic Factors

The post-traumatic fate of neurons and glial cells can be influenced by neurotrophic factors, such as vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), basic fibroblast growth factor (bFGF), and epidermal growth factor (EGF). Administering of these growth factors following traumatic brain injury may enhance the recovery and neurological outcome [145,146].

7.2. Suppression of RhoA GTPase

Recent studies demonstrated that the small GTPase RhoA is essential for mediating the effects of molecules acting against axonal regeneration on damaged myelin and glial scar. The ADP-ribosylation of Rho proteins by the exoenzyme C3-transferrase from Clostridium botulinum may block the downstream signaling of this cascade. As a result of the ADP-ribosylation, the axon regeneration is facilitated [147,148].
Due to the wide variety of cellular functions that C3-transferase has in promoting CNS regeneration, combined therapy using this enzyme and other neuroprotective medications may have an additive impact in the treatment of TBI [149]. Given the parallels between TBI and spinal injuries, it can be presumed that the benefits of C3-transferase treatment seen in spinal injuries could also apply to TBI, although the importance of C3-transferase in experimental models of TBI has not yet been established.

7.3. DNA Vaccine

DNA vaccination is a novel and relatively simple method of inducing an immunological response. Recent studies have suggested that a DNA vaccine encoding the inhibitory epitopes of myelin-associated proteins may be a promising approach to repairing damaged CNS [150,151].

7.4. Protein S100B

The S100B protein is a calcium-binding protein produced by glial cells. The S100B protein has been detected in serum after breaching the BBB following brain injury [152]. It has been shown that the administration of S100B exhibits a dose-dependent dual effect on neurons. In small doses, S100B acts as a neurotrophic neuroprotective factor. However, at high doses, S100B increases neuroinflammation and impairs neuronal survival [153,154].

7.5. Overcoming the Glial Scar

Significant activation of chondroitin sulfate proteoglycans (CSPGs) such as neurocan, phosphacan, versican, and NG2 in the glial scar contributes to impaired axonal regeneration after CNS injury. Administration of CSPG-cleaving enzyme chondroitinase ABC reduces CSPG levels and cavitation at the site of injury within 24 h [155]. Overexpression of chondroitinase ABC in transgenic mice also improved axon regeneration in astrocytic scars [156]. Thus, the administration of inhibitory molecules to the glial scar may represent promising targets for accelerating regeneration in TBI.

7.6. Stem Cell Therapy

Losses of neurons and glia are the main signs of CNS damage. Therefore, the replacement of these cells represents a viable approach to therapy. Injection of mesenchymal stem cells in an acute TBI model reduced the expression of various proinflammatory cytokines and chemokines, such as IL-1β, IL-6, TNF-α, CCL2, CCL11, and CXCL [157]. In addition, recent studies have shown that stem cells improve functional recovery after TBI due to regenerative and neuromodulatory properties [158].
Among various types of stem cells, numerous studies suggested the excellent neuroprotective properties of neural stem cell (NSC)transplantation. By mitigating neuroinflammation and stimulating regenerative processes (i.e., enhancing neurogenesis, angiogenesis, and plasticity), NSCs may support functional recovery after acute TBI [159,160,161].
Guo S. et al. (2019) demonstrated that when administered intranasally, the exosomes derived from mesenchymal stem cells (MSC-Exo) can cross the blood–brain barrier and migrate to the injured area of the spinal cord [162]. MSC-Exo loaded with phosphatase and tensin homolog small interfering RNA (ExoPTEN) significantly enhanced axon growth and neovascularization while reducing microgliosis and astrogliosis. Intranasal therapy with ExoPTEN may also partially improve structural and electrophysiological function and, most importantly, significantly induce functional recovery in rats with complete spinal cord interruption. The results show that intranasal ExoPTEN can be used in a clinical setting to speed up the recovery of patients with spinal cord injuries [162].
Overall, stem cell therapy has been demonstrated as a useful tool that can reduce the effects of TBI. These treatments appear to be safe and have been shown to improve neurological and motor function in TBI patients.

7.7. Nanoparticles

The clinical use of pharmacological treatment in TBI has been hampered by limited possibilities for drug delivery to the brain. In addition, the retention of pharmacological substances in the brain is also limited. Therefore, the use of nanoparticles as drug carriers has received increasing attention in the last few years. Encapsulation of drugs in micro- or nanoparticles appears to be a promising way to ensure sustained and controlled drug delivery in TBI [163]. Both natural and synthetic polymers have been successfully used as drug carriers and depots. The polymers used as carriers share the following common characteristics: biocompatible, biodegradable, generally inert, and capable of attaching to or encapsulating small molecules and proteins [164].
Recently, it has been shown that treatment of a TBI rat model with cerium oxide (CeO2) nanoparticles can significantly reduce brain damage by restoring cognitive abilities and enhancing antioxidant properties [165]. Another study reported that the administration of immunomodulatory nanoparticles markedly improves motor impairment and reduces inflammation and edema in a mouse model of TBI [166].
Recently, the combination of stem cell therapy with nanoparticle delivery gained increasing research attention. Thus, Narouiepour [167] reported that combining human fetal brain-derived stem cells with niosomal nanoparticles can improve functional recovery and reduce neuroinflammation in a TBI model [167]. In addition, Zhu et al. demonstrated the possibility of labeling human stem cells with superparamagnetic iron oxide nanoparticles and tracking them using magnetic resonance imaging (MRI) [168]. A 34-year-old patient in this study had left temporal lobe craniocerebral damage. The exposed neural tissue was removed during emergency surgery, and the cells were cultured in order to select stem cells. The cultured stem cells were then treated by a contrast agent containing superparamagnetic iron oxide nanoparticles. Next, the autologous cultured stem cells were stereotactically injected into the damaged brain area. The MRI signal change one-week post-implantation was correlated with the growth and accumulation of cells surrounding the lesion. During the second and third weeks, the signal around the lesion grew, indicating that the stem cells moved from the main injection sites to the edge of the injured tissue [168].
Currently, due to relatively low reproducibility, poor scalability, and high cost, the use of nanoparticles in TBI treatment is somewhat limited. Therefore, to increase the clinical application of the nanoparticles, the development of scalable, low-cost synthesis is required.

8. Treatment Based on Physical Principles

8.1. Hyperbaric Oxygen Therapy (HBOT)

Currently, hyperbaric oxygen therapy (HBOT) is considered one of the most important clinical treatments for TBI. Harch et al. [169] treated 16 patients with TBI, post-TBI syndrome, and post-traumatic stress disorder (PTSD) utilizing 40 sessions of HBO at 1.5 ATA of oxygen/60 min for 30 days, which significantly improved symptoms, neurological examination results, comprehensive IQ test results, and cognitive function. In addition, Geng et al. [170] demonstrated that HBO can suppress the activation of inflammatory signals, thereby alleviating TBI. In severe TBI, HBOT reduces mortality and improves functional outcomes [171,172].
In several randomized and randomized controlled trials, HBO at 1.5 ATA of oxygen displayed statistically significant symptomatic and cognitive improvements in patients with mild traumatic brain injury and persistent post-concussion syndrome. Positive and negative results were observed at lower and higher doses of oxygen and pressure [173]. Overall, these studies indicate the successful use of HBOT in intensive care for the treatment of patients with TBI.

8.2. Non-Invasive Brain Stimulation

To date, several technologies of non-invasive brain stimulation have been developed. The most common technologies are transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS). Repetitive transcranial magnetic stimulation (rTMS) is a painless, non-invasive, easy-to-administer treatment with few adverse reactions. It has been shown that rTMS has a specific effect on the rehabilitation of patients with TBI [174,175].
Neville et al. [176] conducted a double-blind randomized controlled trial in 36 patients with TBI who were randomly and equally divided into two groups. Ten sessions of high frequency rTMS (10 Hz) over the left dorsolateral pre-frontal cortex (DLPFC) were administered to TBI patients in the treatment group, while pseudostimulation was administered to TBI patients in the sham group. To evaluate immediate and delayed effects, patients completed cognitive evaluations one week prior to, one week following, and three months following rTMS. The findings suggest that rTMS can enhance post-TBI depression and cognitive function [176].
Recently, Dhaliwal etc. [177] has reported that brain stimulation may have a potential impact on the treatment of TBI and that rTMS can be used as an effective treatment for some post-TBI symptoms, such as depression, tinnitus, and neglect.

9. Complementary Medicine

Complementary and integrative medicine approaches, including acupuncture and herbal medicine, are often used to complement the limitations of conventional medicine [178,179], improving efficacy and sometimes reducing side effects even in the treatment of traumatic brain injury [180,181].However, currently available data suggest insufficient evidence to recommend these complementary methods of TBI treatment in clinical practice.

10. Mild TBI Management

The majority of TBIs (up to 90%) are mild [182]. Although some studies reported a relatively low mortality rate of patients with mild TBI [183], these patients remain at increased risk of mortality in the 5 years post-injury [184]. In addition, many survivors of mild TBI experience symptoms for years. For mild TBI, the World Health Organization (WHO) lists the following diagnostic criteria: “(i) one or more of the following: confusion of disorientation, loss of consciousness for ≤30 min, post-traumatic amnesia (PTA) for <24 h and/or other transient neurological abnormalities, such as focal signs, seizure and intracranial lesion not requiring surgery; AND (ii) a Glasgow Coma Scale (GCS) score of 13–15 after 30 min post head injury or later upon presentation for health care. These manifestations of mild TBI must not be due to drugs, alcohol, medications, caused by other injuries or treatment for other injuries (e.g., psychological trauma, language barrier or coexisting medical conditions) or caused by penetrating craniocerebral injury” [185,186].
Many patients with mild TBI are often discharged from the hospital during the first 24 h post-injury provided there are no serious complications or worsening of the status of the patient. Some mild TBI symptoms may appear immediately after the trauma, while others may not appear for hours or days after the injury. Moreover, some patients may remain generally asymptomatic for 1–4 months. Symptoms generally improve over time and many patients feel better within a couple of weeks.
Symptoms of mild TBI are different from person to person. The most common include headache, dizziness, fatigue, sleep disturbances (insomnia, hypersomnia, nightmares), attentional difficulties, memory problems, depression, anxiety, photo- and phono-sensitivity, irritability, and depersonalization [186,187]. The long-term consequences of mild TBI may include cognitive deficit [188], dementia [189], Alzheimer’s disease [190,191], and a high risk of developing neurological disorders, including epilepsy [192] and stroke [193].
The management of mild TBI in many cases could be symptomatic. However, careful consideration of the severity of the initial injury was recommended [194]. While the WHO definition of mild TBI stated above applies to mild TBI, the possibility for a more severe TBI should be assessed and excluded by Mayo criteria [194]. Imaging techniques (MRI, CT scan) should be implemented, especially when suspecting post-traumatic amnesia (PTA), bleeding, or expanding lesions [186]. The proper assessment of individual neurological and neuropsychiatric symptoms should determine individualized treatment. At early stages and in patients with somatic complaints, the intervention should address the treatment of the somatic symptoms. In patients with cognitive or emotional symptoms, neuropsychological help is advised [186,187]. Thus, for example, antidepressants as well as psychotherapy could be administered to patients with depression and anxiety; cognitive behavioral therapy could be effective in reducing concentration and memory difficulties [186]. The overall approach to the treatment of mild TBI should be, therefore, individualized and highly dependent on the severity and character of the injury.

11. Conclusions

TBI is a major global health problem and one of the main priorities in modern critical care. Despite the lack of an effective treatment for TBI recovery, efforts to develop therapeutic strategies for TBI recovery have been continuously undertaken over the past few decades. Today, standard medical and surgical interventions always play a significant role in the emergency care of patients with TBI.
There are several promising areas of research aimed at optimizing neuroresuscitation protocols, developing an evidence base for surgical intervention, and promising neuroprotective pharmacotherapy. Given the heterogeneity of the pathology and the fact that one intervention method cannot affect all involved pathological mechanisms, the importance of personalized medicine and combination therapy in the field of TBI cannot be overestimated.

Author Contributions

Conceptualization, M.S.; investigation, M.S., A.S., M.K., M.A., A.K., E.K., R.K., Z.B. and S.I.; writing—original draft preparation, M.S., A.S., M.K., M.A., A.K., E.K., R.K., Z.B. and S.I.; writing—review and editing, A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Patel, A.; Vieira, M.M.; Abraham, J.; Reid, N.; Tran, T.; Tomecsek, K.; Vissoci, J.R.; Eucker, S.; Gerardo, C.J.; Staton, C.A. Quality of the Development of Traumatic Brain Injury Clinical Practice Guidelines: A Systematic Review. PLoS ONE 2016, 11, e0161554. [Google Scholar] [CrossRef]
  2. Hawryluk, G.W.J.; Rubiano, A.M.; Totten, A.M.; O’Reilly, C.; Ullman, J.S.; Bratton, S.L.; Chesnut, R.; Harris, O.A.; Kissoon, N.; Shutter, L.; et al. Guidelines for the Management of Severe Traumatic Brain Injury: 2020 Update of the Decompressive Craniectomy Recommendations. Neurosurgery 2020, 87, 427–434. [Google Scholar] [CrossRef] [PubMed]
  3. Nichol, A.; French, C.; Little, L.; Haddad, S.; Presneill, J.; Arabi, Y.; Bailey, M.; Cooper, D.J.; Duranteau, J.; Huet, O.; et al. Erythropoietin in traumatic brain injury (EPO-TBI): A double-blind randomised controlled trial. Lancet 2015, 386, 2499–2506. [Google Scholar] [CrossRef]
  4. Wright, D.W.; Yeatts, S.D.; Silbergleit, R.; Palesch, Y.Y.; Hertzberg, V.S.; Frankel, M.; Goldstein, F.C.; Caveney, A.F.; Howlett-Smith, H.; Bengelink, E.M.; et al. Very early administration of progesterone for acute traumatic brain injury. N. Engl. J. Med. 2014, 371, 2457–2466. [Google Scholar] [CrossRef] [PubMed]
  5. Kontos, A.P.; Collins, M.W.; Holland, C.L.; Reeves, V.L.; Edelman, K.; Benso, S.; Schneider, W.; Okonkwo, D. Preliminary Evidence for Improvement in Symptoms, Cognitive, Vestibular, and Oculomotor Outcomes Following Targeted Intervention with Chronic mTBI Patients. Mil. Med. 2018, 183, 333–338. [Google Scholar] [CrossRef] [PubMed]
  6. Carney, N.; Totten, A.M.; O’Reilly, C.; Ullman, J.S.; Hawryluk, G.W.; Bell, M.J.; Bratton, S.L.; Chesnut, R.; Harris, O.A.; Kissoon, N.; et al. Guidelines for the Management of Severe Traumatic Brain Injury, Fourth Edition. Neurosurgery 2017, 80, 6–15. [Google Scholar] [CrossRef]
  7. Kochanek, P.M.; Carney, N.; Adelson, P.D.; Ashwal, S.; Bell, M.J.; Bratton, S.; Carson, S.; Chesnut, R.M.; Ghajar, J.; Goldstein, B.; et al. Guidelines for the acute medical management of severe traumatic brain injury in infants, children, and adolescents--second edition. Pediatr. Crit. Care Med. 2012, 13 (Suppl. S1), S1–S82. [Google Scholar] [CrossRef]
  8. Chesnut, R.M.; Temkin, N.; Carney, N.; Dikmen, S.; Rondina, C.; Videtta, W.; Petroni, G.; Lujan, S.; Pridgeon, J.; Barber, J.; et al. A trial of intracranial-pressure monitoring in traumatic brain injury. N. Engl. J. Med. 2012, 367, 2471–2481. [Google Scholar] [CrossRef] [PubMed]
  9. Kochanek, P.M.; Jackson, T.C.; Ferguson, N.M.; Carlson, S.W.; Simon, D.W.; Brockman, E.C.; Ji, J.; Bayir, H.; Poloyac, S.M.; Wagner, A.K.; et al. Emerging therapies in traumatic brain injury. Semin. Neurol. 2015, 35, 83–100. [Google Scholar] [CrossRef]
  10. Smith, D.H.; Hicks, R.; Povlishock, J.T. Therapy development for diffuse axonal injury. J. Neurotrauma 2013, 30, 307–323. [Google Scholar] [CrossRef]
  11. Mondello, S.; Shear, D.A.; Bramlett, H.M.; Dixon, C.E.; Schmid, K.E.; Dietrich, W.D.; Wang, K.K.; Hayes, R.L.; Glushakova, O.; Catania, M.; et al. Insight into Pre-Clinical Models of Traumatic Brain Injury Using Circulating Brain Damage Biomarkers: Operation Brain Trauma Therapy. J. Neurotrauma 2016, 33, 595–605. [Google Scholar] [CrossRef] [PubMed]
  12. Yang, Z.; Zhu, T.; Mondello, S.; Akel, M.; Wong, A.T.; Kothari, I.M.; Lin, F.; Shear, D.A.; Gilsdorf, J.S.; Leung, L.Y.; et al. Serum-Based Phospho-Neurofilament-Heavy Protein as Theranostic Biomarker in Three Models of Traumatic Brain Injury: An Operation Brain Trauma Therapy Study. J. Neurotrauma 2019, 36, 348–359. [Google Scholar] [CrossRef] [PubMed]
  13. Browning, M.; Shear, D.A.; Bramlett, H.M.; Dixon, C.E.; Mondello, S.; Schmid, K.E.; Poloyac, S.M.; Dietrich, W.D.; Hayes, R.L.; Wang, K.K.; et al. Levetiracetam Treatment in Traumatic Brain Injury: Operation Brain Trauma Therapy. J. Neurotrauma 2016, 33, 581–594. [Google Scholar] [CrossRef] [PubMed]
  14. Jha, R.M.; Molyneaux, B.J.; Jackson, T.C.; Wallisch, J.S.; Park, S.Y.; Poloyac, S.; Vagni, V.A.; Janesko-Feldman, K.L.; Hoshitsuki, K.; Minnigh, M.B.; et al. Glibenclamide Produces Region-Dependent Effects on Cerebral Edema in a Combined Injury Model of Traumatic Brain Injury and Hemorrhagic Shock in Mice. J. Neurotrauma 2018, 35, 2125–2135. [Google Scholar] [CrossRef]
  15. Jha, R.M.; Mondello, S.; Bramlett, H.M.; Dixon, C.E.; Shear, D.A.; Dietrich, W.D.; Wang, K.K.W.; Yang, Z.; Hayes, R.L.; Poloyac, S.M.; et al. Glibenclamide Treatment in Traumatic Brain Injury: Operation Brain Trauma Therapy. J. Neurotrauma 2021, 38, 628–645. [Google Scholar] [CrossRef] [PubMed]
  16. Aghili-Mehrizi, S.; Williams, E.; Yan, S.; Willman, M.; Willman, J.; Lucke-Wold, B. Secondary Mechanisms of Neurotrauma: A Closer Look at the Evidence. Diseases 2022, 10, 30. [Google Scholar] [CrossRef] [PubMed]
  17. Ismail, H.; Shakkour, Z.; Tabet, M.; Abdelhady, S.; Kobaisi, A.; Abedi, R.; Nasrallah, L.; Pintus, G.; Al-Dhaheri, Y.; Mondello, S.; et al. Traumatic Brain Injury: Oxidative Stress and Novel Anti-Oxidants Such as Mitoquinone and Edaravone. Antioxidants 2020, 9, 943. [Google Scholar] [CrossRef] [PubMed]
  18. Ray, S.K.; Dixon, C.E.; Banik, N.L. Molecular mechanisms in the pathogenesis of traumatic brain injury. Histol. Histopathol. 2002, 17, 1137–1152. [Google Scholar] [CrossRef] [PubMed]
  19. Jassam, Y.N.; Izzy, S.; Whalen, M.; McGavern, D.B.; El Khoury, J. Neuroimmunology of Traumatic Brain Injury: Time for a Paradigm Shift. Neuron 2017, 95, 1246–1265. [Google Scholar] [CrossRef]
  20. Kaur, P.; Sharma, S. Recent Advances in Pathophysiology of Traumatic Brain Injury. Curr. Neuropharmacol. 2018, 16, 1224–1238. [Google Scholar] [CrossRef]
  21. Bramlett, H.M.; Dietrich, W.D. Long-Term Consequences of Traumatic Brain Injury: Current Status of Potential Mechanisms of Injury and Neurological Outcomes. J. Neurotrauma 2015, 32, 1834–1848. [Google Scholar] [CrossRef] [PubMed]
  22. Mira, R.G.; Lira, M.; Cerpa, W. Traumatic Brain Injury: Mechanisms of Glial Response. Front. Physiol. 2021, 12, 740939. [Google Scholar] [CrossRef]
  23. Frati, A.; Cerretani, D.; Fiaschi, A.I.; Frati, P.; Gatto, V.; La Russa, R.; Pesce, A.; Pinchi, E.; Santurro, A.; Fraschetti, F.; et al. Diffuse Axonal Injury and Oxidative Stress: A Comprehensive Review. Int. J. Mol. Sci. 2017, 18, 2600. [Google Scholar] [CrossRef] [PubMed]
  24. 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] [PubMed]
  25. Oakes, S.A.; Papa, F.R. The role of endoplasmic reticulum stress in human pathology. Annu. Rev. Pathol. 2015, 10, 173–194. [Google Scholar] [CrossRef]
  26. Chen, X.; Mi, L.; Gu, G.; Gao, X.; Gao, X.; Shi, M.; Chai, Y.; Chen, F.; Yang, W.; Zhang, J. Dysfunctional Endoplasmic Reticulum-Mitochondrion Coupling Is Associated with Endoplasmic Reticulum Stress-Induced Apoptosis and Neurological Deficits in a Rodent Model of Severe Head Injury. J. Neurotrauma 2022, 39, 560–576. [Google Scholar] [CrossRef]
  27. Shi, M.; Chai, Y.; Zhang, J.; Chen, X. Endoplasmic Reticulum Stress-Associated Neuronal Death and Innate Immune Response in Neurological Diseases. Front. Immunol. 2021, 12, 794580. [Google Scholar] [CrossRef]
  28. McAllister, T.W. Neurobiological consequences of traumatic brain injury. Dialogues Clin. Neurosci. 2011, 13, 287–300. [Google Scholar] [CrossRef]
  29. Mietto, B.S.; Mostacada, K.; Martinez, A.M. Neurotrauma and inflammation: CNS and PNS responses. Mediat. Inflamm. 2015, 2015, 251204. [Google Scholar] [CrossRef]
  30. Swadron, S.P.; LeRoux, P.; Smith, W.S.; Weingart, S.D. Emergency neurological life support: Traumatic brain injury. Neurocrit Care 2012, 17 (Suppl. S1), S112–S121. [Google Scholar] [CrossRef]
  31. Osier, N.D.; Carlson, S.W.; DeSana, A.; Dixon, C.E. Chronic Histopathological and Behavioral Outcomes of Experimental Traumatic Brain Injury in Adult Male Animals. J. Neurotrauma 2015, 32, 1861–1882. [Google Scholar] [CrossRef] [PubMed]
  32. Geeraerts, T.; Velly, L.; Abdennour, L.; Asehnoune, K.; Audibert, G.; Bouzat, P.; Bruder, N.; Carrillon, R.; Cottenceau, V.; Cotton, F.; et al. Management of severe traumatic brain injury (first 24hours). Anaesth. Crit. Care Pain. Med. 2018, 37, 171–186. [Google Scholar] [CrossRef] [PubMed]
  33. Feldman, Z.; Kanter, M.J.; Robertson, C.S.; Contant, C.F.; Hayes, C.; Sheinberg, M.A.; Villareal, C.A.; Narayan, R.K.; Grossman, R.G. Effect of head elevation on intracranial pressure, cerebral perfusion pressure, and cerebral blood flow in head-injured patients. J. Neurosurg. 1992, 76, 207–211. [Google Scholar] [CrossRef] [PubMed]
  34. Steidl, C.; Bosel, J.; Suntrup-Krueger, S.; Schonenberger, S.; Al-Suwaidan, F.; Warnecke, T.; Minnerup, J.; Dziewas, R.; Initiative for German Neuro-Intensive Trial, E. Tracheostomy, Extubation, Reintubation: Airway Management Decisions in Intubated Stroke Patients. Cerebrovasc. Dis. 2017, 44, 1–9. [Google Scholar] [CrossRef] [PubMed]
  35. Young, D.; Harrison, D.A.; Cuthbertson, B.H.; Rowan, K.; TracMan, C. Effect of early vs late tracheostomy placement on survival in patients receiving mechanical ventilation: The TracMan randomized trial. JAMA 2013, 309, 2121–2129. [Google Scholar] [CrossRef] [PubMed]
  36. McCredie, V.A.; Adhikari, N.K. Early tracheostomy in critically ill patients: Still too fast. Lancet Respir. Med. 2015, 3, 95–96. [Google Scholar] [CrossRef] [PubMed]
  37. McCredie, V.A.; Alali, A.S.; Scales, D.C.; Adhikari, N.K.; Rubenfeld, G.D.; Cuthbertson, B.H.; Nathens, A.B. Effect of Early Versus Late Tracheostomy or Prolonged Intubation in Critically Ill Patients with Acute Brain Injury: A Systematic Review and Meta-Analysis. Neurocrit Care 2017, 26, 14–25. [Google Scholar] [CrossRef]
  38. Hyde, G.A.; Savage, S.A.; Zarzaur, B.L.; Hart-Hyde, J.E.; Schaefer, C.B.; Croce, M.A.; Fabian, T.C. Early tracheostomy in trauma patients saves time and money. Injury 2015, 46, 110–114. [Google Scholar] [CrossRef]
  39. Alali, A.S.; Scales, D.C.; Fowler, R.A.; Mainprize, T.G.; Ray, J.G.; Kiss, A.; de Mestral, C.; Nathens, A.B. Tracheostomy timing in traumatic brain injury: A propensity-matched cohort study. J. Trauma Acute Care Surg. 2014, 76, 70–76; discussion 76–78. [Google Scholar] [CrossRef]
  40. Bosel, J.; Schiller, P.; Hacke, W.; Steiner, T. Benefits of early tracheostomy in ventilated stroke patients? Current evidence and study protocol of the randomized pilot trial SETPOINT (Stroke-related Early Tracheostomy vs. Prolonged Orotracheal Intubation in Neurocritical care Trial). Int. J. Stroke 2012, 7, 173–182. [Google Scholar] [CrossRef]
  41. Bosel, J.; Schiller, P.; Hook, Y.; Andes, M.; Neumann, J.O.; Poli, S.; Amiri, H.; Schonenberger, S.; Peng, Z.; Unterberg, A.; et al. Stroke-related Early Tracheostomy versus Prolonged Orotracheal Intubation in Neurocritical Care Trial (SETPOINT): A randomized pilot trial. Stroke 2013, 44, 21–28. [Google Scholar] [CrossRef] [PubMed]
  42. Shekhar, S.; Singh, R.B.; De, R.R.; Singh, R.; Akhileshwar; Kumar, N. Early versus Late Tracheostomy in Patients with Acute Brain Injury: Importance of SET Score. Anesth. Essays Res. 2022, 16, 7–11. [Google Scholar] [CrossRef] [PubMed]
  43. Grubb, R.L., Jr.; Raichle, M.E.; Eichling, J.O.; Ter-Pogossian, M.M. The effects of changes in PaCO2 on cerebral blood volume, blood flow, and vascular mean transit time. Stroke 1974, 5, 630–639. [Google Scholar] [CrossRef] [PubMed]
  44. Darby, J.M.; Yonas, H.; Marion, D.W.; Latchaw, R.E. Local “inverse steal” induced by hyperventilation in head injury. Neurosurgery 1988, 23, 84–88. [Google Scholar] [CrossRef] [PubMed]
  45. Jovanovic, B.; Milan, Z.; Markovic-Denic, L.; Djuric, O.; Radinovic, K.; Doklestic, K.; Velickovic, J.; Ivancevic, N.; Gregoric, P.; Pandurovic, M.; et al. Risk factors for ventilator-associated pneumonia in patients with severe traumatic brain injury in a Serbian trauma centre. Int. J. Infect. Dis. 2015, 38, 46–51. [Google Scholar] [CrossRef]
  46. Esnault, P.; Nguyen, C.; Bordes, J.; D’Aranda, E.; Montcriol, A.; Contargyris, C.; Cotte, J.; Goutorbe, P.; Joubert, C.; Dagain, A.; et al. Early-Onset Ventilator-Associated Pneumonia in Patients with Severe Traumatic Brain Injury: Incidence, Risk Factors, and Consequences in Cerebral Oxygenation and Outcome. Neurocrit Care 2017, 27, 187–198. [Google Scholar] [CrossRef] [PubMed]
  47. Gamberini, L.; Giugni, A.; Ranieri, S.; Meconi, T.; Coniglio, C.; Gordini, G.; Bardi, T. Early-Onset Ventilator-Associated Pneumonia in Severe Traumatic Brain Injury: Is There a Relationship with Prehospital Airway Management? J. Emerg. Med. 2019, 56, 657–665. [Google Scholar] [CrossRef]
  48. Torbic, H.; Forni, A.A.; Anger, K.E.; Degrado, J.R.; Greenwood, B.C. Use of antiepileptics for seizure prophylaxis after traumatic brain injury. Am. J. Health Syst. Pharm. 2013, 70, 759–766. [Google Scholar] [CrossRef]
  49. Zafar, S.N.; Khan, A.A.; Ghauri, A.A.; Shamim, M.S. Phenytoin versus Leviteracetam for seizure prophylaxis after brain injury—A meta analysis. BMC Neurol. 2012, 12, 30. [Google Scholar] [CrossRef]
  50. Chou, A.; Krukowski, K.; Jopson, T.; Zhu, P.J.; Costa-Mattioli, M.; Walter, P.; Rosi, S. Inhibition of the integrated stress response reverses cognitive deficits after traumatic brain injury. Proc. Natl. Acad. Sci. USA 2017, 114, E6420–E6426. [Google Scholar] [CrossRef]
  51. Oddo, M.; Crippa, I.A.; Mehta, S.; Menon, D.; Payen, J.F.; Taccone, F.S.; Citerio, G. Optimizing sedation in patients with acute brain injury. Crit. Care 2016, 20, 128. [Google Scholar] [CrossRef] [PubMed]
  52. Sharshar, T.; Citerio, G.; Andrews, P.J.; Chieregato, A.; Latronico, N.; Menon, D.K.; Puybasset, L.; Sandroni, C.; Stevens, R.D. Neurological examination of critically ill patients: A pragmatic approach. Report of an ESICM expert panel. Intensive Care Med. 2014, 40, 484–495. [Google Scholar] [CrossRef] [PubMed]
  53. Bayir, H.; Adelson, P.D.; Wisniewski, S.R.; Shore, P.; Lai, Y.; Brown, D.; Janesko-Feldman, K.L.; Kagan, V.E.; Kochanek, P.M. Therapeutic hypothermia preserves antioxidant defenses after severe traumatic brain injury in infants and children. Crit. Care Med. 2009, 37, 689–695. [Google Scholar] [CrossRef] [PubMed]
  54. Andrews, P.J.; Sinclair, H.L.; Rodriguez, A.; Harris, B.; Rhodes, J.; Watson, H.; Murray, G. Therapeutic hypothermia to reduce intracranial pressure after traumatic brain injury: The Eurotherm3235 RCT. Health Technol. Assess. 2018, 22, 1–134. [Google Scholar] [CrossRef] [PubMed]
  55. Lewis, S.R.; Evans, D.J.; Butler, A.R.; Schofield-Robinson, O.J.; Alderson, P. Hypothermia for traumatic brain injury. Cochrane Database Syst. Rev. 2017, 9, CD001048. [Google Scholar] [CrossRef] [PubMed]
  56. Cooper, D.J.; Nichol, A.D.; Bailey, M.; Bernard, S.; Cameron, P.A.; Pili-Floury, S.; Forbes, A.; Gantner, D.; Higgins, A.M.; Huet, O.; et al. Effect of Early Sustained Prophylactic Hypothermia on Neurologic Outcomes Among Patients With Severe Traumatic Brain Injury: The POLAR Randomized Clinical Trial. JAMA 2018, 320, 2211–2220. [Google Scholar] [CrossRef] [PubMed]
  57. Polderman, K.H. Induced hypothermia and fever control for prevention and treatment of neurological injuries. Lancet 2008, 371, 1955–1969. [Google Scholar] [CrossRef]
  58. Badjatia, N. Hypothermia in neurocritical care. Neurosurg. Clin. N. Am. 2013, 24, 457–467. [Google Scholar] [CrossRef]
  59. Vella, M.A.; Crandall, M.L.; Patel, M.B. Acute Management of Traumatic Brain Injury. Surg. Clin. N. Am. 2017, 97, 1015–1030. [Google Scholar] [CrossRef]
  60. Puccio, A.M.; Fischer, M.R.; Jankowitz, B.T.; Yonas, H.; Darby, J.M.; Okonkwo, D.O. Induced normothermia attenuates intracranial hypertension and reduces fever burden after severe traumatic brain injury. Neurocrit Care 2009, 11, 82–87. [Google Scholar] [CrossRef]
  61. Klauber, M.R.; Marshall, L.F.; Luerssen, T.G.; Frankowski, R.; Tabaddor, K.; Eisenberg, H.M. Determinants of head injury mortality: Importance of the low risk patient. Neurosurgery 1989, 24, 31–36. [Google Scholar] [CrossRef] [PubMed]
  62. Fuller, G.; Hasler, R.M.; Mealing, N.; Lawrence, T.; Woodford, M.; Juni, P.; Lecky, F. The association between admission systolic blood pressure and mortality in significant traumatic brain injury: A multi-centre cohort study. Injury 2014, 45, 612–617. [Google Scholar] [CrossRef]
  63. Pigott, A.; Rudloff, E. Traumatic Brain Injury-A Review of Intravenous Fluid Therapy. Front. Vet. Sci. 2021, 8, 643800. [Google Scholar] [CrossRef]
  64. Alvis-Miranda, H.R.; Castellar-Leones, S.M.; Moscote-Salazar, L.R. Intravenous Fluid Therapy in Traumatic Brain Injury and Decompressive Craniectomy. Bull. Emerg. Trauma 2014, 2, 3–14. [Google Scholar] [PubMed]
  65. Dash, H.H.; Chavali, S. Management of traumatic brain injury patients. Korean J. Anesthesiol. 2018, 71, 12–21. [Google Scholar] [CrossRef] [PubMed]
  66. White, H.; Cook, D.; Venkatesh, B. The use of hypertonic saline for treating intracranial hypertension after traumatic brain injury. Anesth. Analg. 2006, 102, 1836–1846. [Google Scholar] [CrossRef] [PubMed]
  67. Weed, L.H.; McKibben, P.S. Experimental alteration of brain bulk. Am. J. Physiol. 1919, 48, 531–558. [Google Scholar] [CrossRef]
  68. Rudloff, E.; Hopper, K. Crystalloid and Colloid Compositions and Their Impact. Front. Vet. Sci. 2021, 8, 639848. [Google Scholar] [CrossRef] [PubMed]
  69. Wakai, A.; McCabe, A.; Roberts, I.; Schierhout, G. Mannitol for acute traumatic brain injury. Cochrane Database Syst. Rev. 2013, 8, CD001049. [Google Scholar] [CrossRef]
  70. Maas, A.I.; Dearden, M.; Teasdale, G.M.; Braakman, R.; Cohadon, F.; Iannotti, F.; Karimi, A.; Lapierre, F.; Murray, G.; Ohman, J.; et al. EBIC-guidelines for management of severe head injury in adults. European Brain Injury Consortium. Acta Neurochir. 1997, 139, 286–294. [Google Scholar] [CrossRef]
  71. The Brain Trauma Foundation; The American Association of Neurological Surgeons. The Joint Section on Neurotrauma and Critical Care. Computed tomography scan features. J. Neurotrauma 2000, 17, 597–627. [Google Scholar] [CrossRef] [PubMed]
  72. Dorman, H.R.; Sondheimer, J.H.; Cadnapaphornchai, P. Mannitol-induced acute renal failure. Medicine 1990, 69, 153–159. [Google Scholar] [CrossRef] [PubMed]
  73. Roberts, I.; Schierhout, G.; Wakai, A. Mannitol for acute traumatic brain injury. Cochrane Database Syst. Rev. 2003, 2, CD001049. [Google Scholar] [CrossRef] [PubMed]
  74. SAFE Study Investigators; Australian and New Zealand Intensive Care Society Clinical Trials Group; Australian Red Cross Blood Service; George Institute for International Health; Myburgh, J.; Cooper, D.J.; Finfer, S.; Bellomo, R.; Norton, R.; Bishop, N.; et al. Saline or albumin for fluid resuscitation in patients with traumatic brain injury. N. Engl. J. Med. 2007, 357, 874–884. [Google Scholar] [CrossRef]
  75. Li, N.; Zhao, W.G.; Zhang, W.F. Acute kidney injury in patients with severe traumatic brain injury: Implementation of the acute kidney injury network stage system. Neurocrit Care 2011, 14, 377–381. [Google Scholar] [CrossRef] [PubMed]
  76. Perel, P.; Roberts, I.; Ker, K. Colloids versus crystalloids for fluid resuscitation in critically ill patients. Cochrane Database Syst. Rev. 2013, 6, CD000567. [Google Scholar] [CrossRef] [PubMed]
  77. Roberts, I.; Shakur-Still, H.; Aeron-Thomas, A.; Beaumont, D.; Belli, A.; Brenner, A.; Cargill, M.; Chaudhri, R.; Douglas, N.; Frimley, L.; et al. Tranexamic acid to reduce head injury death in people with traumatic brain injury: The CRASH-3 international RCT. Health Technol. Assess. 2021, 25, 1–76. [Google Scholar] [CrossRef] [PubMed]
  78. Yokobori, S.; Yatabe, T.; Kondo, Y.; Kinoshita, K.; Kosaku Kinoshita for the Japan Resuscitation Council (JRC) Neuroresuscitation Task Force and the Guidelines Editorial Committee. Efficacy and safety of tranexamic acid administration in traumatic brain injury patients: A systematic review and meta-analysis. J. Intensive Care 2020, 8, 46. [Google Scholar] [CrossRef] [PubMed]
  79. Bossers, S.M.; Loer, S.A.; Bloemers, F.W.; Den Hartog, D.; Van Lieshout, E.M.M.; Hoogerwerf, N.; van der Naalt, J.; Absalom, A.R.; Peerdeman, S.M.; Schwarte, L.A.; et al. Association Between Prehospital Tranexamic Acid Administration and Outcomes of Severe Traumatic Brain Injury. JAMA Neurol. 2021, 78, 338–345. [Google Scholar] [CrossRef] [PubMed]
  80. Huang, H.; Xin, M.; Wu, X.; Liu, J.; Zhang, W.; Yang, K.; Zhang, J. The efficacy of tranexamic acid treatment with different time and doses for traumatic brain injury: A systematic review and meta-analysis. Thromb. J. 2022, 20, 79. [Google Scholar] [CrossRef]
  81. Abecassis, I.J.; Young, C.C.; Caldwell, D.J.; Feroze, A.H.; Williams, J.R.; Meyer, R.M.; Kellogg, R.T.; Bonow, R.H.; Chesnut, R.M. The Kempe incision for decompressive craniectomy, craniotomy, and cranioplasty in traumatic brain injury and stroke. J. Neurosurg. 2021, 135, 1807–1816. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, D.; Xue, Q.; Chen, J.; Dong, Y.; Hou, L.; Jiang, Y.; Wang, J. Decompressive craniectomy in the management of intracranial hypertension after traumatic brain injury: A systematic review and meta-analysis. Sci. Rep. 2017, 7, 8800. [Google Scholar] [CrossRef]
  83. Iaccarino, C.; Kolias, A.G.; Roumy, L.G.; Fountas, K.; Adeleye, A.O. Cranioplasty Following Decompressive Craniectomy. Front. Neurol. 2019, 10, 1357. [Google Scholar] [CrossRef] [PubMed]
  84. Chandra, V.V.R.; Mowliswara Prasad, B.C.; Banavath, H.N.; Chandrasekhar Reddy, K. Cisternostomy versus Decompressive Craniectomy for the Management of Traumatic Brain Injury: A Randomized Controlled Trial. World Neurosurg. 2022, 162, e58–e64. [Google Scholar] [CrossRef]
  85. Parthiban, J.; Sundaramahalingam, S.; Rao, J.B.; Nannaware, V.P.; Rathwa, V.N.; Nasre, V.Y.; Prahlad, S.T. Basal Cisternostomy—A Microsurgical Cerebro Spinal Fluid Let Out Procedure and Treatment Option in the Management of Traumatic Brain Injury. Analysis of 40 Consecutive Head Injury Patients Operated with and without Bone Flap Replacement Following Cisternostomy in a Tertiary Care Centre in India. Neurol. India 2021, 69, 328–333. [Google Scholar] [CrossRef]
  86. Giammattei, L.; Starnoni, D.; Messerer, M.; Daniel, R.T. Basal Cisternostomy for Severe TBI: Surgical Technique and Cadaveric Dissection. Front. Surg. 2022, 9, 915818. [Google Scholar] [CrossRef]
  87. 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 10,008 adults with clinically significant head injury (MRC CRASH trial): Randomised placebo-controlled trial. Lancet 2004, 364, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  88. Aloizos, S.; Evodia, E.; Gourgiotis, S.; Isaia, E.C.; Seretis, C.; Baltopoulos, G.J. Neuroprotective Effects of Erythropoietin in Patients with Severe Closed Brain Injury. Turk. Neurosurg. 2015, 25, 552–558. [Google Scholar] [CrossRef]
  89. Li, Z.M.; Xiao, Y.L.; Zhu, J.X.; Geng, F.Y.; Guo, C.J.; Chong, Z.L.; Wang, L.X. Recombinant human erythropoietin improves functional recovery in patients with severe traumatic brain injury: A randomized, double blind and controlled clinical trial. Clin. Neurol. Neurosurg. 2016, 150, 80–83. [Google Scholar] [CrossRef]
  90. Lee, J.; Cho, Y.; Choi, K.S.; Kim, W.; Jang, B.H.; Shin, H.; Ahn, C.; Lim, T.H.; Yi, H.J. Efficacy and safety of erythropoietin in patients with traumatic brain injury: A systematic review and meta-analysis. Am. J. Emerg. Med. 2019, 37, 1101–1107. [Google Scholar] [CrossRef]
  91. Skrifvars, M.B.; Bailey, M.; French, C.; Presneill, J.; Nichol, A.; Little, L.; Duranteau, J.; Huet, O.; Haddad, S.; Arabi, Y.; et al. Erythropoietin in patients with traumatic brain injury and extracranial injury-A post hoc analysis of the erythropoietin traumatic brain injury trial. J. Trauma Acute Care Surg. 2017, 83, 449–456. [Google Scholar] [CrossRef] [PubMed]
  92. Mizoguchi, K.; Yokoo, H.; Yoshida, M.; Tanaka, T.; Tanaka, M. Amantadine increases the extracellular dopamine levels in the striatum by re-uptake inhibition and by N-methyl-D-aspartate antagonism. Brain Res. 1994, 662, 255–258. [Google Scholar] [CrossRef] [PubMed]
  93. Sawyer, E.; Mauro, L.S.; Ohlinger, M.J. Amantadine enhancement of arousal and cognition after traumatic brain injury. Ann. Pharmacother. 2008, 42, 247–252. [Google Scholar] [CrossRef] [PubMed]
  94. Giacino, J.T.; Whyte, J.; Bagiella, E.; Kalmar, K.; Childs, N.; Khademi, A.; Eifert, B.; Long, D.; Katz, D.I.; Cho, S.; et al. Placebo-controlled trial of amantadine for severe traumatic brain injury. N. Engl. J. Med. 2012, 366, 819–826. [Google Scholar] [CrossRef] [PubMed]
  95. Spritzer, S.D.; Kinney, C.L.; Condie, J.; Wellik, K.E.; Hoffman-Snyder, C.R.; Wingerchuk, D.M.; Demaerschalk, B.M. Amantadine for patients with severe traumatic brain injury: A critically appraised topic. Neurologist 2015, 19, 61–64. [Google Scholar] [CrossRef] [PubMed]
  96. Ellis, E.F.; Dodson, L.Y.; Police, R.J. Restoration of cerebrovascular responsiveness to hyperventilation by the oxygen radical scavenger n-acetylcysteine following experimental traumatic brain injury. J. Neurosurg. 1991, 75, 774–779. [Google Scholar] [CrossRef] [PubMed]
  97. Xiong, Y.; Peterson, P.L.; Lee, C.P. Effect of N-acetylcysteine on mitochondrial function following traumatic brain injury in rats. J. Neurotrauma 1999, 16, 1067–1082. [Google Scholar] [CrossRef]
  98. Chen, G.; Shi, J.; Hu, Z.; Hang, C. Inhibitory effect on cerebral inflammatory response following traumatic brain injury in rats: A potential neuroprotective mechanism of N-acetylcysteine. Mediat. Inflamm. 2008, 2008, 716458. [Google Scholar] [CrossRef] [PubMed]
  99. Hoffer, M.E.; Balaban, C.; Slade, M.D.; Tsao, J.W.; Hoffer, B. Amelioration of acute sequelae of blast induced mild traumatic brain injury by N-acetyl cysteine: A double-blind, placebo controlled study. PLoS ONE 2013, 8, e54163. [Google Scholar] [CrossRef]
  100. Haber, M.; James, J.; Kim, J.; Sangobowale, M.; Irizarry, R.; Ho, J.; Nikulina, E.; Grin’kina, N.M.; Ramadani, A.; Hartman, I.; et al. Minocycline plus N-acteylcysteine induces remyelination, synergistically protects oligodendrocytes and modifies neuroinflammation in a rat model of mild traumatic brain injury. J. Cereb. Blood Flow. Metab. 2018, 38, 1312–1326. [Google Scholar] [CrossRef]
  101. Zhang, L.; Xiao, H.; Yu, X.; Deng, Y. Minocycline attenuates neurological impairment and regulates iron metabolism in a rat model of traumatic brain injury. Arch. Biochem. Biophys. 2020, 682, 108302. [Google Scholar] [CrossRef] [PubMed]
  102. Kadir, A.; Andreasen, N.; Almkvist, O.; Wall, A.; Forsberg, A.; Engler, H.; Hagman, G.; Larksater, M.; Winblad, B.; Zetterberg, H.; et al. Effect of phenserine treatment on brain functional activity and amyloid in Alzheimer’s disease. Ann. Neurol. 2008, 63, 621–631. [Google Scholar] [CrossRef] [PubMed]
  103. Poole, N.A.; Agrawal, N. Cholinomimetic agents and neurocognitive impairment following head injury: A systematic review. Brain Inj. 2008, 22, 519–534. [Google Scholar] [CrossRef] [PubMed]
  104. Hsueh, S.C.; Lecca, D.; Greig, N.H.; Wang, J.Y.; Selman, W.; Hoffer, B.J.; Miller, J.P.; Chiang, Y.H. (-)-Phenserine Ameliorates Contusion Volume, Neuroinflammation, and Behavioral Impairments Induced by Traumatic Brain Injury in Mice. Cell Transplant. 2019, 28, 1183–1196. [Google Scholar] [CrossRef] [PubMed]
  105. Lecca, D.; Bader, M.; Tweedie, D.; Hoffman, A.F.; Jung, Y.J.; Hsueh, S.C.; Hoffer, B.J.; Becker, R.E.; Pick, C.G.; Lupica, C.R.; et al. (-)-Phenserine and the prevention of pre-programmed cell death and neuroinflammation in mild traumatic brain injury and Alzheimer’s disease challenged mice. Neurobiol. Dis. 2019, 130, 104528. [Google Scholar] [CrossRef] [PubMed]
  106. Langham, J.; Goldfrad, C.; Teasdale, G.; Shaw, D.; Rowan, K. Calcium channel blockers for acute traumatic brain injury. Cochrane Database Syst. Rev. 2003, 2003, CD000565. [Google Scholar] [CrossRef] [PubMed]
  107. Vergouwen, M.D.; Vermeulen, M.; Roos, Y.B. Effect of nimodipine on outcome in patients with traumatic subarachnoid haemorrhage: A systematic review. Lancet Neurol. 2006, 5, 1029–1032. [Google Scholar] [CrossRef] [PubMed]
  108. Verweij, B.H.; Muizelaar, J.P.; Vinas, F.C.; Peterson, P.L.; Xiong, Y.; Lee, C.P. Improvement in mitochondrial dysfunction as a new surrogate efficiency measure for preclinical trials: Dose-response and time-window profiles for administration of the calcium channel blocker Ziconotide in experimental brain injury. J. Neurosurg. 2000, 93, 829–834. [Google Scholar] [CrossRef]
  109. Berman, R.F.; Verweij, B.H.; Muizelaar, J.P. Neurobehavioral protection by the neuronal calcium channel blocker ziconotide in a model of traumatic diffuse brain injury in rats. J. Neurosurg. 2000, 93, 821–828. [Google Scholar] [CrossRef]
  110. Lee, L.L.; Galo, E.; Lyeth, B.G.; Muizelaar, J.P.; Berman, R.F. Neuroprotection in the rat lateral fluid percussion model of traumatic brain injury by SNX-185, an N-type voltage-gated calcium channel blocker. Exp. Neurol. 2004, 190, 70–78. [Google Scholar] [CrossRef]
  111. Kulbe, J.R.; Singh, I.N.; Wang, J.A.; Cebak, J.E.; Hall, E.D. Continuous Infusion of Phenelzine, Cyclosporine A, or Their Combination: Evaluation of Mitochondrial Bioenergetics, Oxidative Damage, and Cytoskeletal Degradation following Severe Controlled Cortical Impact Traumatic Brain Injury in Rats. J. Neurotrauma 2018, 35, 1280–1293. [Google Scholar] [CrossRef] [PubMed]
  112. Mazzeo, A.T.; Brophy, G.M.; Gilman, C.B.; Alves, O.L.; Robles, J.R.; Hayes, R.L.; Povlishock, J.T.; Bullock, M.R. Safety and tolerability of cyclosporin a in severe traumatic brain injury patients: Results from a prospective randomized trial. J. Neurotrauma 2009, 26, 2195–2206. [Google Scholar] [CrossRef] [PubMed]
  113. Alali, A.S.; McCredie, V.A.; Golan, E.; Shah, P.S.; Nathens, A.B. Beta blockers for acute traumatic brain injury: A systematic review and meta-analysis. Neurocrit Care 2014, 20, 514–523. [Google Scholar] [CrossRef]
  114. Cotton, B.A.; Snodgrass, K.B.; Fleming, S.B.; Carpenter, R.O.; Kemp, C.D.; Arbogast, P.G.; Morris, J.A., Jr. Beta-blocker exposure is associated with improved survival after severe traumatic brain injury. J. Trauma 2007, 62, 26–35. [Google Scholar] [CrossRef] [PubMed]
  115. Ko, A.; Harada, M.Y.; Barmparas, G.; Thomsen, G.M.; Alban, R.F.; Bloom, M.B.; Chung, R.; Melo, N.; Margulies, D.R.; Ley, E.J. Early propranolol after traumatic brain injury is associated with lower mortality. J. Trauma Acute Care Surg. 2016, 80, 637–642. [Google Scholar] [CrossRef] [PubMed]
  116. Ley, E.J.; Leonard, S.D.; Barmparas, G.; Dhillon, N.K.; Inaba, K.; Salim, A.; O’Bosky, K.R.; Tatum, D.; Azmi, H.; Ball, C.G.; et al. Beta blockers in critically ill patients with traumatic brain injury: Results from a multicenter, prospective, observational American Association for the Surgery of Trauma study. J. Trauma Acute Care Surg. 2018, 84, 234–244. [Google Scholar] [CrossRef]
  117. Mohseni, S.; Talving, P.; Wallin, G.; Ljungqvist, O.; Riddez, L. Preinjury beta-blockade is protective in isolated severe traumatic brain injury. J. Trauma Acute Care Surg. 2014, 76, 804–808. [Google Scholar] [CrossRef]
  118. Alexander, R.W.; Davis, J.N.; Lefkowitz, R.J. Direct identification and characterisation of beta-adrenergic receptors in rat brain. Nature 1975, 258, 437–440. [Google Scholar] [CrossRef]
  119. MacKenzie, E.T.; McCulloch, J.; Harper, A.M. Influence of endogenous norepinephrine on cerebral blood flow and metabolism. Am. J. Physiol. 1976, 231, 489–494. [Google Scholar] [CrossRef]
  120. Heffernan, D.S.; Inaba, K.; Arbabi, S.; Cotton, B.A. Sympathetic hyperactivity after traumatic brain injury and the role of beta-blocker therapy. J. Trauma 2010, 69, 1602–1609. [Google Scholar] [CrossRef]
  121. Kemp, C.D.; Johnson, J.C.; Riordan, W.P.; Cotton, B.A. How we die: The impact of nonneurologic organ dysfunction after severe traumatic brain injury. Am. Surg. 2008, 74, 866–872. [Google Scholar] [CrossRef] [PubMed]
  122. Khalili, H.; Ahl, R.; Paydar, S.; Sjolin, G.; Cao, Y.; Abdolrahimzadeh Fard, H.; Niakan, A.; Hanna, K.; Joseph, B.; Mohseni, S. Beta-Blocker Therapy in Severe Traumatic Brain Injury: A Prospective Randomized Controlled Trial. World J. Surg. 2020, 44, 1844–1853. [Google Scholar] [CrossRef] [PubMed]
  123. Ding, H.; Liao, L.; Zheng, X.; Wang, Q.; Liu, Z.; Xu, G.; Li, X.; Liu, L. beta-Blockers for traumatic brain injury: A systematic review and meta-analysis. J. Trauma Acute Care Surg. 2021, 90, 1077–1085. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, J.; Gallagher, D.; DeVito, L.M.; Cancino, G.I.; Tsui, D.; He, L.; Keller, G.M.; Frankland, P.W.; Kaplan, D.R.; Miller, F.D. Metformin activates an atypical PKC-CBP pathway to promote neurogenesis and enhance spatial memory formation. Cell Stem Cell 2012, 11, 23–35. [Google Scholar] [CrossRef] [PubMed]
  125. Afshari, K.; Dehdashtian, A.; Haddadi, N.S.; Haj-Mirzaian, A.; Iranmehr, A.; Ebrahimi, M.A.; Tavangar, S.M.; Faghir-Ghanesefat, H.; Mohammadi, F.; Rahimi, N.; et al. Anti-inflammatory effects of Metformin improve the neuropathic pain and locomotor activity in spinal cord injured rats: Introduction of an alternative therapy. Spinal Cord. 2018, 56, 1032–1041. [Google Scholar] [CrossRef] [PubMed]
  126. Kothari, V.; Galdo, J.A.; Mathews, S.T. Hypoglycemic agents and potential anti-inflammatory activity. J. Inflamm. Res. 2016, 9, 27–38. [Google Scholar] [CrossRef] [PubMed]
  127. Cameron, A.R.; Morrison, V.L.; Levin, D.; Mohan, M.; Forteath, C.; Beall, C.; McNeilly, A.D.; Balfour, D.J.; Savinko, T.; Wong, A.K.; et al. Anti-Inflammatory Effects of Metformin Irrespective of Diabetes Status. Circ. Res. 2016, 119, 652–665. [Google Scholar] [CrossRef] [PubMed]
  128. Hill, J.L.; Kobori, N.; Zhao, J.; Rozas, N.S.; Hylin, M.J.; Moore, A.N.; Dash, P.K. Traumatic brain injury decreases AMP-activated protein kinase activity and pharmacological enhancement of its activity improves cognitive outcome. J. Neurochem. 2016, 139, 106–119. [Google Scholar] [CrossRef] [PubMed]
  129. Tao, L.; Li, D.; Liu, H.; Jiang, F.; Xu, Y.; Cao, Y.; Gao, R.; Chen, G. Neuroprotective effects of metformin on traumatic brain injury in rats associated with NF-kappaB and MAPK signaling pathway. Brain Res. Bull. 2018, 140, 154–161. [Google Scholar] [CrossRef]
  130. Wang, H.; Zheng, Z.; Han, W.; Yuan, Y.; Li, Y.; Zhou, K.; Wang, Q.; Xie, L.; Xu, K.; Zhang, H.; et al. Metformin Promotes Axon Regeneration after Spinal Cord Injury through Inhibiting Oxidative Stress and Stabilizing Microtubule. Oxid. Med. Cell Longev. 2020, 2020, 9741369. [Google Scholar] [CrossRef]
  131. DiBona, V.L.; Shah, M.K.; Krause, K.J.; Zhu, W.; Voglewede, M.M.; Smith, D.M.; Crockett, D.P.; Zhang, H. Metformin reduces neuroinflammation and improves cognitive functions after traumatic brain injury. Neurosci. Res. 2021, 172, 99–109. [Google Scholar] [CrossRef]
  132. Fiani, B.; Covarrubias, C.; Wong, A.; Doan, T.; Reardon, T.; Nikolaidis, D.; Sarno, E. Cerebrolysin for stroke, neurodegeneration, and traumatic brain injury: Review of the literature and outcomes. Neurol. Sci. 2021, 42, 1345–1353. [Google Scholar] [CrossRef] [PubMed]
  133. Wong, G.K.; Zhu, X.L.; Poon, W.S. Beneficial effect of cerebrolysin on moderate and severe head injury patients: Result of a cohort study. Acta Neurochir. Suppl. 2005, 95, 59–60. [Google Scholar] [CrossRef]
  134. Chen, C.C.; Wei, S.T.; Tsaia, S.C.; Chen, X.X.; Cho, D.Y. Cerebrolysin enhances cognitive recovery of mild traumatic brain injury patients: Double-blind, placebo-controlled, randomized study. Br. J. Neurosurg. 2013, 27, 803–807. [Google Scholar] [CrossRef]
  135. Alvarez, X.A.; Sampedro, C.; Figueroa, J.; Tellado, I.; Gonzalez, A.; Garcia-Fantini, M.; Cacabelos, R.; Muresanu, D.; Moessler, H. Reductions in qEEG slowing over 1 year and after treatment with Cerebrolysin in patients with moderate-severe traumatic brain injury. J. Neural Transm. 2008, 115, 683–692. [Google Scholar] [CrossRef] [PubMed]
  136. Alvarez, X.A.; Sampedro, C.; Perez, P.; Laredo, M.; Couceiro, V.; Hernandez, A.; Figueroa, J.; Varela, M.; Arias, D.; Corzo, L.; et al. Positive effects of cerebrolysin on electroencephalogram slowing, cognition and clinical outcome in patients with postacute traumatic brain injury: An exploratory study. Int. Clin. Psychopharmacol. 2003, 18, 271–278. [Google Scholar] [CrossRef]
  137. Onose, G.; Muresanu, D.F.; Ciurea, A.V.; Daia Chendreanu, C.; Mihaescu, A.S.; Mardare, D.C.; Andone, I.; Spanu, A.; Popescu, C.; Dumitrescu, A.; et al. Neuroprotective and consequent neurorehabilitative clinical outcomes, in patients treated with the pleiotropic drug cerebrolysin. J. Med. Life 2009, 2, 350–360. [Google Scholar]
  138. Ghaffarpasand, F.; Torabi, S.; Rasti, A.; Niakan, M.H.; Aghabaklou, S.; Pakzad, F.; Beheshtian, M.S.; Tabrizi, R. Effects of cerebrolysin on functional outcome of patients with traumatic brain injury: A systematic review and meta-analysis. Neuropsychiatr. Dis. Treat. 2019, 15, 127–135. [Google Scholar] [CrossRef]
  139. Lucena, L.L.N.; Briones, M.V.A. Effect of Cerebrolysin in severe traumatic brain injury: A multi-center, retrospective cohort study. Clin. Neurol. Neurosurg. 2022, 216, 107216. [Google Scholar] [CrossRef] [PubMed]
  140. Jamall, O.A.; Feeney, C.; Zaw-Linn, J.; Malik, A.; Niemi, M.E.; Tenorio-Jimenez, C.; Ham, T.E.; Jilka, S.R.; Jenkins, P.O.; Scott, G.; et al. Prevalence and correlates of vitamin D deficiency in adults after traumatic brain injury. Clin. Endocrinol. 2016, 85, 636–644. [Google Scholar] [CrossRef]
  141. Lee, P.; Nair, P.; Eisman, J.A.; Center, J.R. Vitamin D deficiency in the intensive care unit: An invisible accomplice to morbidity and mortality? Intensive Care Med. 2009, 35, 2028–2032. [Google Scholar] [CrossRef] [PubMed]
  142. Viglianti, E.M.; Zajic, P.; Iwashyna, T.J.; Amrein, K. Neither vitamin D levels nor supplementation are associated with the development of persistent critical illness: A retrospective cohort analysis. Crit. Care Resusc. 2019, 21, 39–44. [Google Scholar] [CrossRef] [PubMed]
  143. Williams, S.; Heuberger, R. Outcomes of Vitamin D Supplementation in Adults Who are Deficient and Critically Ill: A Review of the Literature. Am. J. Ther. 2016, 23, e1890–e1902. [Google Scholar] [CrossRef] [PubMed]
  144. Arabi, S.M.; Sedaghat, A.; Ehsaei, M.R.; Safarian, M.; Ranjbar, G.; Rezaee, H.; Rezvani, R.; Tabesh, H.; Norouzy, A. Efficacy of high-dose versus low-dose vitamin D supplementation on serum levels of inflammatory factors and mortality rate in severe traumatic brain injury patients: Study protocol for a randomized placebo-controlled trial. Trials 2020, 21, 685. [Google Scholar] [CrossRef] [PubMed]
  145. Wu, H.; Lu, D.; Jiang, H.; Xiong, Y.; Qu, C.; Li, B.; Mahmood, A.; Zhou, D.; Chopp, M. Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury. J. Neurotrauma 2008, 25, 130–139. [Google Scholar] [CrossRef] [PubMed]
  146. Sun, D.; Bullock, M.R.; McGinn, M.J.; Zhou, Z.; Altememi, N.; Hagood, S.; Hamm, R.; Colello, R.J. Basic fibroblast growth factor-enhanced neurogenesis contributes to cognitive recovery in rats following traumatic brain injury. Exp. Neurol. 2009, 216, 56–65. [Google Scholar] [CrossRef] [PubMed]
  147. Auer, M.; Schweigreiter, R.; Hausott, B.; Thongrong, S.; Holtje, M.; Just, I.; Bandtlow, C.; Klimaschewski, L. Rho-independent stimulation of axon outgrowth and activation of the ERK and Akt signaling pathways by C3 transferase in sensory neurons. Front. Cell Neurosci. 2012, 6, 43. [Google Scholar] [CrossRef] [PubMed]
  148. Auer, M.; Allodi, I.; Barham, M.; Udina, E.; Neiss, W.F.; Navarro, X.; Klimaschewski, L. C3 exoenzyme lacks effects on peripheral axon regeneration in vivo. J. Peripher. Nerv. Syst. 2013, 18, 30–36. [Google Scholar] [CrossRef]
  149. McKerracher, L.; Guertin, P. Rho as a target to promote repair: Translation to clinical studies with cethrin. Curr. Pharm. Des. 2013, 19, 4400–4410. [Google Scholar] [CrossRef]
  150. Zhang, Y.; Ang, B.T.; Xiao, Z.C.; Ng, I. DNA vaccination against neurite growth inhibitors to enhance functional recovery following traumatic brain injury. Acta Neurochir. Suppl. 2008, 102, 347–351. [Google Scholar] [CrossRef]
  151. Xu, G.; Nie, D.Y.; Chen, J.T.; Wang, C.Y.; Yu, F.G.; Sun, L.; Luo, X.G.; Ahmed, S.; David, S.; Xiao, Z.C. Recombinant DNA vaccine encoding multiple domains related to inhibition of neurite outgrowth: A potential strategy for axonal regeneration. J. Neurochem. 2004, 91, 1018–1023. [Google Scholar] [CrossRef]
  152. Blyth, B.J.; Farhavar, A.; Gee, C.; Hawthorn, B.; He, H.; Nayak, A.; Stocklein, V.; Bazarian, J.J. Validation of serum markers for blood-brain barrier disruption in traumatic brain injury. J. Neurotrauma 2009, 26, 1497–1507. [Google Scholar] [CrossRef] [PubMed]
  153. Thelin, E.P.; Nelson, D.W.; Bellander, B.M. A review of the clinical utility of serum S100B protein levels in the assessment of traumatic brain injury. Acta Neurochir. 2017, 159, 209–225. [Google Scholar] [CrossRef] [PubMed]
  154. Sanchez-Pena, P.; Pereira, A.R.; Sourour, N.A.; Biondi, A.; Lejean, L.; Colonne, C.; Boch, A.L.; Al Hawari, M.; Abdennour, L.; Puybasset, L. S100B as an additional prognostic marker in subarachnoid aneurysmal hemorrhage. Crit. Care Med. 2008, 36, 2267–2273. [Google Scholar] [CrossRef] [PubMed]
  155. Lin, R.; Kwok, J.C.; Crespo, D.; Fawcett, J.W. Chondroitinase ABC has a long-lasting effect on chondroitin sulphate glycosaminoglycan content in the injured rat brain. J. Neurochem. 2008, 104, 400–408. [Google Scholar] [CrossRef] [PubMed]
  156. Cafferty, W.B.; Yang, S.H.; Duffy, P.J.; Li, S.; Strittmatter, S.M. Functional axonal regeneration through astrocytic scar genetically modified to digest chondroitin sulfate proteoglycans. J. Neurosci. 2007, 27, 2176–2185. [Google Scholar] [CrossRef] [PubMed]
  157. Galindo, L.T.; Filippo, T.R.; Semedo, P.; Ariza, C.B.; Moreira, C.M.; Camara, N.O.; Porcionatto, M.A. Mesenchymal stem cell therapy modulates the inflammatory response in experimental traumatic brain injury. Neurol. Res. Int. 2011, 2011, 564089. [Google Scholar] [CrossRef]
  158. Luo, M.L.; Pan, L.; Wang, L.; Wang, H.Y.; Li, S.; Long, Z.Y.; Zeng, L.; Liu, Y. Transplantation of NSCs Promotes the Recovery of Cognitive Functions by Regulating Neurotransmitters in Rats with Traumatic Brain Injury. Neurochem. Res. 2019, 44, 2765–2775. [Google Scholar] [CrossRef] [PubMed]
  159. Moshayedi, P.; Nih, L.R.; Llorente, I.L.; Berg, A.R.; Cinkornpumin, J.; Lowry, W.E.; Segura, T.; Carmichael, S.T. Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain. Biomaterials 2016, 105, 145–155. [Google Scholar] [CrossRef]
  160. Blaya, M.O.; Tsoulfas, P.; Bramlett, H.M.; Dietrich, W.D. Neural progenitor cell transplantation promotes neuroprotection, enhances hippocampal neurogenesis, and improves cognitive outcomes after traumatic brain injury. Exp. Neurol. 2015, 264, 67–81. [Google Scholar] [CrossRef]
  161. Gao, J.; Grill, R.J.; Dunn, T.J.; Bedi, S.; Labastida, J.A.; Hetz, R.A.; Xue, H.; Thonhoff, J.R.; DeWitt, D.S.; Prough, D.S.; et al. Human Neural Stem Cell Transplantation-Mediated Alteration of Microglial/Macrophage Phenotypes after Traumatic Brain Injury. Cell Transplant. 2016, 25, 1863–1877. [Google Scholar] [CrossRef] [PubMed]
  162. Guo, S.; Perets, N.; Betzer, O.; Ben-Shaul, S.; Sheinin, A.; Michaelevski, I.; Popovtzer, R.; Offen, D.; Levenberg, S. Intranasal Delivery of Mesenchymal Stem Cell Derived Exosomes Loaded with Phosphatase and Tensin Homolog siRNA Repairs Complete Spinal Cord Injury. ACS Nano 2019, 13, 10015–10028. [Google Scholar] [CrossRef] [PubMed]
  163. Bharadwaj, V.N.; Nguyen, D.T.; Kodibagkar, V.D.; Stabenfeldt, S.E. Nanoparticle-Based Therapeutics for Brain Injury. Adv. Healthc. Mater. 2018, 7, 1700668. [Google Scholar] [CrossRef]
  164. Orive, G.; Anitua, E.; Pedraz, J.L.; Emerich, D.F. Biomaterials for promoting brain protection, repair and regeneration. Nat. Rev. Neurosci. 2009, 10, 682–692. [Google Scholar] [CrossRef]
  165. Bailey, Z.S.; Nilson, E.; Bates, J.A.; Oyalowo, A.; Hockey, K.S.; Sajja, V.; Thorpe, C.; Rogers, H.; Dunn, B.; Frey, A.S.; et al. Cerium Oxide Nanoparticles Improve Outcome after In Vitro and In Vivo Mild Traumatic Brain Injury. J. Neurotrauma 2020, 37, 1452–1462. [Google Scholar] [CrossRef] [PubMed]
  166. Sharma, S.; Ifergan, I.; Kurz, J.E.; Linsenmeier, R.A.; Xu, D.; Cooper, J.G.; Miller, S.D.; Kessler, J.A. Intravenous Immunomodulatory Nanoparticle Treatment for Traumatic Brain Injury. Ann. Neurol. 2020, 87, 442–455. [Google Scholar] [CrossRef]
  167. Narouiepour, A.; Ebrahimzadeh-Bideskan, A.; Rajabzadeh, G.; Gorji, A.; Negah, S.S. Neural stem cell therapy in conjunction with curcumin loaded in niosomal nanoparticles enhanced recovery from traumatic brain injury. Sci. Rep. 2022, 12, 3572. [Google Scholar] [CrossRef] [PubMed]
  168. Zhu, J.; Zhou, L.; XingWu, F. Tracking neural stem cells in patients with brain trauma. N. Engl. J. Med. 2006, 355, 2376–2378. [Google Scholar] [CrossRef]
  169. Harch, P.G.; Andrews, S.R.; Fogarty, E.F.; Amen, D.; Pezzullo, J.C.; Lucarini, J.; Aubrey, C.; Taylor, D.V.; Staab, P.K.; Van Meter, K.W. A phase I study of low-pressure hyperbaric oxygen therapy for blast-induced post-concussion syndrome and post-traumatic stress disorder. J. Neurotrauma 2012, 29, 168–185. [Google Scholar] [CrossRef]
  170. Geng, F.; Ma, Y.; Xing, T.; Zhuang, X.; Zhu, J.; Yao, L. Effects of Hyperbaric Oxygen Therapy on Inflammasome Signaling after Traumatic Brain Injury. Neuroimmunomodulation 2016, 23, 122–129. [Google Scholar] [CrossRef]
  171. Lv, L.Q.; Hou, L.J.; Yu, M.K.; Ding, X.H.; Qi, X.Q.; Lu, Y.C. Hyperbaric oxygen therapy in the management of paroxysmal sympathetic hyperactivity after severe traumatic brain injury: A report of 6 cases. Arch. Phys. Med. Rehabil. 2011, 92, 1515–1518. [Google Scholar] [CrossRef] [PubMed]
  172. Prakash, A.; Parelkar, S.V.; Oak, S.N.; Gupta, R.K.; Sanghvi, B.V.; Bachani, M.; Patil, R. Role of hyperbaric oxygen therapy in severe head injury in children. J. Pediatr. Neurosci. 2012, 7, 4–8. [Google Scholar] [CrossRef] [PubMed]
  173. Harch, P.G. Systematic Review and Dosage Analysis: Hyperbaric Oxygen Therapy Efficacy in Mild Traumatic Brain Injury Persistent Postconcussion Syndrome. Front. Neurol. 2022, 13, 815056. [Google Scholar] [CrossRef] [PubMed]
  174. Kreuzer, P.M.; Landgrebe, M.; Frank, E.; Langguth, B. Repetitive transcranial magnetic stimulation for the treatment of chronic tinnitus after traumatic brain injury: A case study. J. Head. Trauma Rehabil. 2013, 28, 386–389. [Google Scholar] [CrossRef] [PubMed]
  175. Li, S.; Zaninotto, A.L.; Neville, I.S.; Paiva, W.S.; Nunn, D.; Fregni, F. Clinical utility of brain stimulation modalities following traumatic brain injury: Current evidence. Neuropsychiatr. Dis. Treat. 2015, 11, 1573–1586. [Google Scholar] [CrossRef] [PubMed]
  176. Neville, I.S.; Hayashi, C.Y.; El Hajj, S.A.; Zaninotto, A.L.; Sabino, J.P.; Sousa, L.M., Jr.; Nagumo, M.M.; Brunoni, A.R.; Shieh, B.D.; Amorim, R.L.; et al. Repetitive Transcranial Magnetic Stimulation (rTMS) for the cognitive rehabilitation of traumatic brain injury (TBI) victims: Study protocol for a randomized controlled trial. Trials 2015, 16, 440. [Google Scholar] [CrossRef] [PubMed]
  177. Dhaliwal, S.K.; Meek, B.P.; Modirrousta, M.M. Non-Invasive Brain Stimulation for the Treatment of Symptoms Following Traumatic Brain Injury. Front. Psychiatry 2015, 6, 119. [Google Scholar] [CrossRef] [PubMed]
  178. Galgano, M.; Toshkezi, G.; Qiu, X.; Russell, T.; Chin, L.; Zhao, L.R. Traumatic Brain Injury: Current Treatment Strategies and Future Endeavors. Cell Transplant. 2017, 26, 1118–1130. [Google Scholar] [CrossRef] [PubMed]
  179. Polich, G.; Iaccarino, M.A.; Kaptchuk, T.J.; Morales-Quezada, L.; Zafonte, R. Placebo Effects in Traumatic Brain Injury. J. Neurotrauma 2018, 35, 1205–1212. [Google Scholar] [CrossRef]
  180. Tan, L.; Zeng, L.; Wang, N.; Deng, M.; Chen, Y.; Ma, T.; Zhang, L.; Xu, Z. Acupuncture to Promote Recovery of Disorder of Consciousness after Traumatic Brain Injury: A Systematic Review and Meta-Analysis. Evid. Based Complement. Altern. Med. 2019, 2019, 5190515. [Google Scholar] [CrossRef]
  181. Gau, B.S.; Yang, H.L.; Huang, S.J.; Lou, M.F. The use of complementary and alternative medicine for patients with traumatic brain injury in Taiwan. BMC Complement. Altern. Med. 2012, 12, 211. [Google Scholar] [CrossRef] [PubMed]
  182. Kay, A.; Teasdale, G. Head injury in the United Kingdom. World J. Surg. 2001, 25, 1210–1220. [Google Scholar] [CrossRef] [PubMed]
  183. af Geijerstam, J.L.; Britton, M. Mild head injury—Mortality and complication rate: Meta-analysis of findings in a systematic literature review. Acta Neurochir. 2003, 145, 843–850; discussion 850. [Google Scholar] [CrossRef] [PubMed]
  184. Sercy, E.; Orlando, A.; Carrick, M.; Lieser, M.; Madayag, R.; Vasquez, D.; Tanner, A., 2nd; Rubin, B.; Bar-Or, D. Long-term mortality and causes of death among patients with mild traumatic brain injury: A 5-year multicenter study. Brain Inj. 2020, 34, 556–566. [Google Scholar] [CrossRef] [PubMed]
  185. Carroll, L.J.; Cassidy, J.D.; Holm, L.; Kraus, J.; Coronado, V.G. Methodological issues and research recommendations for mild traumatic brain injury: The WHO Collaborating Centre Task Force on Mild Traumatic Brain Injury. J. Rehabil. Med. 2004, 36, 113–125. [Google Scholar] [CrossRef] [PubMed]
  186. van Gils, A.; Stone, J.; Welch, K.; Davidson, L.R.; Kerslake, D.; Caesar, D.; McWhirter, L.; Carson, A. Management of mild traumatic brain injury. Pract. Neurol. 2020, 20, 213–221. [Google Scholar] [CrossRef] [PubMed]
  187. Kushner, D. Mild traumatic brain injury: Toward understanding manifestations and treatment. Arch. Intern. Med. 1998, 158, 1617–1624. [Google Scholar] [CrossRef] [PubMed]
  188. McInnes, K.; Friesen, C.L.; MacKenzie, D.E.; Westwood, D.A.; Boe, S.G. Mild Traumatic Brain Injury (mTBI) and chronic cognitive impairment: A scoping review. PLoS ONE 2017, 12, e0174847. [Google Scholar] [CrossRef] [PubMed]
  189. Snowden, T.M.; Hinde, A.K.; Reid, H.M.O.; Christie, B.R. Does Mild Traumatic Brain Injury Increase the Risk for Dementia? A Systematic Review and Meta-Analysis. J. Alzheimers Dis. 2020, 78, 757–775. [Google Scholar] [CrossRef]
  190. Nemetz, P.N.; Leibson, C.; Naessens, J.M.; Beard, M.; Kokmen, E.; Annegers, J.F.; Kurland, L.T. Traumatic brain injury and time to onset of Alzheimer’s disease: A population-based study. Am. J. Epidemiol. 1999, 149, 32–40. [Google Scholar] [CrossRef]
  191. Graham, A.; Livingston, G.; Purnell, L.; Huntley, J. Mild Traumatic Brain Injuries and Future Risk of Developing Alzheimer’s Disease: Systematic Review and Meta-Analysis. J. Alzheimers Dis. 2022, 87, 969–979. [Google Scholar] [CrossRef] [PubMed]
  192. Annegers, J.F.; Coan, S.P. The risks of epilepsy after traumatic brain injury. Seizure 2000, 9, 453–457. [Google Scholar] [CrossRef] [PubMed]
  193. Turner, G.M.; McMullan, C.; Aiyegbusi, O.L.; Bem, D.; Marshall, T.; Calvert, M.; Mant, J.; Belli, A. Stroke risk following traumatic brain injury: Systematic review and meta-analysis. Int. J. Stroke 2021, 16, 370–384. [Google Scholar] [CrossRef] [PubMed]
  194. Malec, J.F.; Brown, A.W.; Leibson, C.L.; Flaada, J.T.; Mandrekar, J.N.; Diehl, N.N.; Perkins, P.K. The mayo classification system for traumatic brain injury severity. J. Neurotrauma 2007, 24, 1417–1424. [Google Scholar] [CrossRef]
Table 1. Overview of the modern approach to the treatment of traumatic brain injury.
Table 1. Overview of the modern approach to the treatment of traumatic brain injury.
Strategies of TBI ManagementSpecific Methods of TBI Treatment
    Emergency care of TBIHead position
Airway management
Tracheostomy
Hyperventilation
Ventilator-associated pneumonia
Prevention of seizures
Sedation and induced coma
Hypothermia
Blood pressure and cerebral perfusion pressure
Fluid management
Tranexamic acid
    Surgical interventions for TBIDecompressive craniectomy
Kempe hemispherectomy incision
Cisternostomy
    Pharmacological therapy of TBICorticosteroids
Progesterone
Erythropoietin
Amantadine
N-acetylcysteine
Minocycline
Phenserine
Calcium channel blockers
Antioxidants
Beta-blockers
Metformin
Cerebrolysin
Vitamin D
    Regenerative treatmentsNeurotrophic factors
Suppression of RhoA GTPase
DNA vaccine
Protein S100B
Overcoming the glial scar
Stem cell therapy
Nanoparticles
    Treatment based on physical principlesHyperbaric oxygen therapy
Non-invasive brain stimulation
    Complementary therapyPhytotherapy
Acupuncture
    Mild TBI management
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Syzdykbayev, M.; Kazymov, M.; Aubakirov, M.; Kurmangazina, A.; Kairkhanov, E.; Kazangapov, R.; Bryzhakhina, Z.; Imangazinova, S.; Sheinin, A. A Modern Approach to the Treatment of Traumatic Brain Injury. Medicines 2024, 11, 10. https://doi.org/10.3390/medicines11050010

AMA Style

Syzdykbayev M, Kazymov M, Aubakirov M, Kurmangazina A, Kairkhanov E, Kazangapov R, Bryzhakhina Z, Imangazinova S, Sheinin A. A Modern Approach to the Treatment of Traumatic Brain Injury. Medicines. 2024; 11(5):10. https://doi.org/10.3390/medicines11050010

Chicago/Turabian Style

Syzdykbayev, Marat, Maksut Kazymov, Marat Aubakirov, Aigul Kurmangazina, Ernar Kairkhanov, Rustem Kazangapov, Zhanna Bryzhakhina, Saule Imangazinova, and Anton Sheinin. 2024. "A Modern Approach to the Treatment of Traumatic Brain Injury" Medicines 11, no. 5: 10. https://doi.org/10.3390/medicines11050010

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