Next Article in Journal
Metabolomic Profiling of Mice with Tacrolimus-Induced Nephrotoxicity: Carnitine Deficiency in Renal Tissue
Previous Article in Journal
Revisiting Treatment of Metastatic Urothelial Cancer: Where Do Cisplatin and Platinum Ineligibility Criteria Stand?
Previous Article in Special Issue
The Relationship between Injury Characteristics and Post-Traumatic Recovery after Diffuse Axonal Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiology-Based Management of Secondary Injuries and Insults in TBI

by
Leonardo de Macedo Filho
1,*,
Luisa F. Figueredo
2,
Gustavo Adolfo Villegas-Gomez
3,
Matthew Arthur
2,
Maria Camila Pedraza-Ciro
4,
Henrique Martins
5,
Joaquim Kanawati Neto
5,
Gregory J. Hawryluk
6 and
Robson Luís Oliveira Amorim
5,7
1
Neurosurgery Department, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
2
Department of Psychiatry, NYU Langone Health, New York, NY 10016, USA
3
Faculty of Medicine, Universidad de Los Andes, Bogota 111711, Colombia
4
Faculty of Medicine, Universidad El Bosque, Bogota 11001, Colombia
5
Postgraduate Program in Health Sciences, Federal University of Amazonas, Manaus 69067-005, Brazil
6
Department of Neurosurgery, Cleveland Clinic and Akron General Hospital, Fairlawn, OH 44307, USA
7
Division of Neurological Surgery, Hospital das Clinicas, University of São Paulo School of Medicine, São Paulo 05508-070, Brazil
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(3), 520; https://doi.org/10.3390/biomedicines12030520
Submission received: 8 November 2023 / Revised: 11 January 2024 / Accepted: 26 January 2024 / Published: 26 February 2024
(This article belongs to the Special Issue Traumatic CNS Injury: From Bench to Bedside)

Abstract

:
Traumatic Brain Injury (TBI) remains a leading cause of morbidity and mortality among all ages; despite the advances, understanding pathophysiological responses after TBI is still complex, involving multiple mechanisms. Previous reviews have focused on potential targets; however, the research on potential targets has continuously grown in the last five years, bringing even more alternatives and elucidating previous mechanisms. Knowing the key and updated pathophysiology concepts is vital for adequate management and better outcomes. This article reviews the underlying molecular mechanisms, the latest updates, and future directions for pathophysiology-based TBI management.

1. Introduction

Traumatic brain injury (TBI) is a leading cause of disability and death among children and young adults, with an incidence of approximately 1.7 million per year in the USA, resulting in 52,000 deaths [1]. Survivors of the initial impact must still contend with the consequences of trauma, as not all injury occurs at the time of impact. The primary injury results from forces applied to the head and involve direct structural damage to the brain. This triggers a cascade of events leading to neurological damage that evolves secondary injury [2]. Several external brain insults, both intracranial and systemic, may complicate and worsen the secondary injury [3].
As was pointed out by Ng et al., current therapeutic strategies focus on preventing secondary injury through early surgical intervention, multiparameter monitoring, and targeted treatment in the intensive care setting [2,4]. Adherence to guidelines for managing severe TBI is associated with better outcomes [5]. However, treatment should be individualized to address underlying mechanisms following TBI and the role of secondary injury in recovery. This work focuses on the underlying pathophysiological mechanisms and their related treatments to systematize the thought process for TBI management, highlighting the role. Importantly, although this article focuses on the mechanisms of excitotoxicity, neuroinflammation and oxidative stress, and blood-brain barrier disruption in TBI, there are other mechanisms central to the pathogenesis of TBI, such as changes in energy metabolism and axonal injury. These topics, although critical, are beyond the scope of this review and deserve detailed analysis in future research.

2. Materials and Methods

We conducted a literature search of PUBMED, SCOPUS, and Google Scholar from 1980 to 2024 using search terms related to the pathophysiology and treatment of traumatic brain injury on 8 January 2024. The searched keywords were ((Secondary TBI) OR (Secondary Brain Injury) OR (Secondary Traumatic Brain Injury)) AND pathophysiology, retrieving 3043 full-text results. This review (Figure 1) aims to concisely evaluate the pathophysiology of TBI and provide guidance for understanding current and emerging treatment modalities. Recommendations in this review are based on the guidelines released by the Brain Trauma Foundation in 2016 [6].
Each section on the pathophysiological basis is followed by a discussion of the specific treatment evolved and its current status. Treatments with multiple action mechanisms are primarily discussed in the section with a more experimental or clinical background. We also searched the clinicaltrials.gov website to evaluate any ongoing clinical trials (status: “recruiting patients”, “not yet recruiting”, or “active, not recruiting”).

3. Literature Review

3.1. Spectrum of TBI Secondary Injuries

3.1.1. Mild Traumatic Brain Injury (mTBI)

mTBI, commonly known as concussion, is characterized by a range of subtle yet significant biochemical and cellular changes [7]. These changes primarily include temporary dysfunction in neuronal connectivity and metabolism [1]. Key features often involve minor axonal disruptions, alterations in ion channels leading to ionic fluxes, and imbalances in neurotransmitters [8]. While the structural integrity of the brain may largely remain intact, these molecular and cellular disturbances can result in symptoms such as headache, dizziness, and short-term memory disturbances [8]. Despite the transient nature of these symptoms, a subset of patients might experience prolonged post-concussive syndromes, wherein symptoms persist for weeks or months [8]. This phenomenon underscores the need for a nuanced understanding of mTBI’s secondary effects, particularly in the context of repeated injuries and their cumulative impact [8].

3.1.2. Severe Traumatic Brain Injury

Severe TBI is marked by more profound and immediate secondary injuries that can be life-threatening and lead to long-term disabilities. The hallmark of severe TBI includes cerebral edema (swelling of the brain), increased intracranial pressure, and significant axonal injury [9]. These injuries can disrupt the blood-brain barrier, leading to inflammatory responses and oxidative stress, which further exacerbate brain damage. Secondary insults in severe TBI may also encompass hemorrhagic lesions, hypoxia, and ischemia [10], contributing to a complex cascade of neurodegeneration. The consequences of these injuries are often severe, manifesting as long-term cognitive, behavioral, and motor impairments [10]. Management of severe TBI is multifaceted, involving acute medical interventions to stabilize the patient and extensive rehabilitation programs to aid recovery and improve quality of life.

3.1.3. Subdural Hematoma (SDH)

SDH occurs when blood collects between the dura mater and the arachnoid layer of the brain [11]. This accumulation of blood leads to increased intracranial pressure, which can cause significant secondary brain injury [11]. Patients with SDH often experience a progressive decline in consciousness, headache, and hemiparesis [11]. The secondary injuries in SDH can include brain herniation, ischemic injury due to compression of blood vessels, and hypoxic injury from reduced blood flow [12]. The management of SDH focuses on surgical intervention to evacuate the hematoma and relieve pressure, followed by monitoring for potential complications like rebleeding or seizures [12].

3.1.4. Subarachnoid Hemorrhage (SAH)

SAH involves bleeding into the subarachnoid space, the area between the arachnoid membrane and the pia mater surrounding the brain [13]. This type of hemorrhage is often caused by the rupture of cerebral aneurysms [13]. The primary concern in SAH is the risk of rebleeding and the development of cerebral vasospasm, which can lead to ischemic injury [14]. Secondary injuries in SAH also include hydrocephalus and elevated intracranial pressure [14]. The management of SAH is complex, often requiring nimble balancing of maintaining cerebral perfusion while minimizing the risk of rebleeding and managing vasospasms, often through medication and surgical interventions [14].

3.1.5. Contusions

Cerebral contusions are essentially bruises on the brain tissue, occurring typically at the site of impact or, in some cases, at a location opposite to the point of impact (coup-contrecoup injury) [15]. These injuries result from the brain striking the inner surface of the skull [16]. Contusions are characterized by localized bleeding and swelling in the brain tissue [15]. The swelling, or edema, can lead to an increase in intracranial pressure, potentially causing a reduction in cerebral blood flow and oxygen supply [17]. In severe cases, this can escalate into a herniation syndrome, where brain tissue is displaced from its normal position [17]. The management of contusions may involve monitoring for changes in neurological status and intracranial pressure, and in some cases, surgical intervention is required to alleviate pressure [17]. Rehabilitation is often necessary to address cognitive and motor deficits resulting from the injury [18].

3.1.6. Diffuse Axonal Injury (DAI)

Diffuse Axonal Injury is a form of TBI that involves widespread damage to the brain’s white matter [19]. This injury occurs when the brain rapidly shifts inside the skull, typically as a result of high-velocity impacts such as those seen in car accidents or falls from significant heights [19]. The shearing forces during the impact disrupt the axons, leading to a breakdown in neuronal communication [20]. The hallmark of DAI is a disruption of the normal signaling and connectivity within the brain, which can lead to a variety of symptoms, ranging from subtle cognitive impairments to prolonged coma [20]. Diagnosis is often challenging due to the microscopic nature of the injury and may require advanced imaging techniques like Diffusion Tensor Imaging (DTI) [21]. The treatment for DAI is largely supportive, focusing on managing symptoms and preventing secondary injuries [21]. Rehabilitation plays a critical role in the recovery process, with a focus on maximizing functional independence and cognitive recovery.

3.1.7. Penetrating Brain Injuries

Penetrating brain injuries occur when an object, such as a bullet or sharp instrument, breaches the skull and enters the brain tissue [22]. These injuries are particularly severe due to the direct damage to brain tissue and the high risk of infection and inflammation [22]. The trajectory and speed of the penetrating object play a significant role in the extent of the injury. Immediate concerns include bleeding, increased intracranial pressure, and the risk of secondary injuries due to brain swelling and herniation [23]. Surgical intervention is often necessary to remove foreign objects, repair damaged tissue, and reduce intracranial pressure [23]. Antibiotic therapy is critical to prevent or treat infections. Long-term rehabilitation is required to address physical, cognitive, and psychological impacts [24]. Due to the complex nature of these injuries, outcomes can vary widely, with some individuals experiencing significant long-term disabilities.

3.2. Pathophysiology of Secondary Brain Injury

Secondary traumatic brain injury is the neurological damage that evolves due to the primary injury (Figure 2). Within the first few hours following the injury, a series of events may trigger a cascade of secondary metabolic, inflammatory, and ischemic insults that can exacerbate the primary neuronal injury. These events are associated with the activation of genes that lead to the transcription of proteins and enzymes involved in the release of excitatory amino acids, the production of free radicals and inflammatory cytokines, alterations in ion flux across membranes, upregulation of neuroprotective cascades, and the induction of programmed cell death [25].
Alterations in cellular metabolism and signaling pathways (Figure 3) can lead to the depletion of energy reserves. As a result, neuronal cells may no longer be able to maintain depolarization of their membranes, causing alterations in ion fluxes and osmotic swelling of the cells. Therefore, following TBI, it is crucial to recognize, prevent, and treat secondary injury to ensure neuronal survival.

3.3. Sequelae from the Primary Impact

3.3.1. Cellular Events

Excitotoxicity and Calcium

Severe TBI causes an abrupt increase in extracellular excitatory amino acids such as glutamate and aspartate [2]. Evidence shows a 40% decline in the expression of astrocytic sodium-dependent glutamate transporters GLAST (EAAT1) and GLT-1 (EAAT2) within 24 h following TBI, leading to a significant decrease in the resorption of glutamate [2]. These excitatory amino acids activate N-methyl-D-Aspartate (NMDA) and non-NMDA receptors, leading to cell membrane depolarization and the influx of sodium (Na2+), potassium (K+), and calcium (Ca2+). High intracellular calcium concentrations activate phospholipase and calpain enzymes that alter membrane and cytoskeletal integrity, eventually resulting in neuronal cell damage and death.
Magnesium (Mg2+) can regulate excitotoxic processes by blocking NMDA receptors and calcium channels. However, Mg2+ concentration decreases after TBI and persists for at least four days after the impact, and its deficiency has been associated with poor neurological outcomes [26]; increasing extracellular magnesium concentration improves the recovery of hippocampal neuronal high-energy phosphates and accelerates regional cerebral flow to the ischemic brain area [27].
Cortical spreading depression (CSD) is a slowly propagating wave of depolarization of neurons and glial cells, followed by a sustained suppression of spontaneous neuronal activity, accompanied by complex and variable changes in vascular caliber, blood flow and energy metabolism [28]. Excitotoxicity is complicated when cortical spreading depression (CSD) occurs [3], a depolarization wave in cerebral gray matter that propagates across the brain [28]. It can break down ion homeostasis, facilitating the release of excitatory amino acids. CSD is associated with poor outcomes in TBI patients [28]. In humans, microdialysis studies demonstrate an increased lactate-pyruvate ratio without consistent evidence of ischemia, and this finding is strongly correlated with outcome [29]. CSD and/or mitochondrial dysfunction may underlie this marker of disturbed metabolism since they are associated with lactate accumulation [25,30].

Specific Treatment

Until this point, statins have been proven to have the potential to protect cultured neurons from excitotoxic death caused by the glutamate receptor agonist NMDA [31]. Interestingly, when evaluating TBI patients with prolonged disorders of consciousness, the use of amantadine, an assumed NMDA antagonist and dopamine agonist, accelerates functional recovery [32]
Multiple clinical trials in acute TBI targeting glutamate and specifically its NMDA receptor have shown contradictory results to demonstrate any beneficial effect [33,34,35], as well as treatment with nimodipine (a calcium channel blocker) [36]. Some authors argue that blocking synaptic transmission mediated by NMDA receptors hinders neuronal survival [34].
Despite promising experimental studies with the use of intravenous magnesium sulfate, its clinical efficiency has yet to be proven magnesium sulfate therapy is effective in the treatment of patients with TBI [37,38].
Regarding CSD, clinical management can focus on controlling factors that increase its incidence and duration, such as systemic hypotension, pyrexia, hypoxia, and low plasma glucose [28].

Free Radicals and Oxidative Stress

Free radicals are by-products of energy metabolism in cells and play a role in vascular tone and immune function. Signaling pathways can increase free radical formation after TBI. Calcium activates pathways that release free radicals from mitochondria and increase nitric oxide (NO) production through inducible NO synthase (iNOS) [39]. Nicotinamide adenine dinucleotide (NADPH) oxidase produces reactive oxygen species (ROS) within the first hour after trauma [40]. ROS increases affinity to NO, forming peroxynitrites (ONOO-) [40], which destroy the cytoskeleton, cell membranes, and DNA [41]. NO inhibits cytochrome c oxidase, causing mitochondrial disruption and cell death [42].
NO formed by vascular endothelium (endothelial nitric oxide synthase—eNOS) exerts beneficial effects after TBI, including vasodilation and increased cerebral blood flow [42]. Peroxynitrites are the leading cause of oxidative stress, as indicated by increased nitrotyrosine in TBI patients with poor outcomes [43]. ONOO- inhibits potassium channels, increasing vascular tone and impairing vascular reactivity [42]. Iron compounds from hemoglobin degradation form reactive free radical oxidants, altering synaptic function and contributing to posttraumatic seizures [44].
  • Specific treatment
Citicoline has multiple neuroprotective mechanisms, including inhibition of oxidative stress and apoptotic pathways [45]. However, a large randomized controlled trial (RCT) showed no benefit in TBI patients [46].
Statins up-regulate eNOS expression and inhibit inducible NO, interleukin1 β (IL1 β), and tumor necrosis factor α (TNF α) [46]. They reduce post-traumatic hypoperfusion and rebound hyperemia [46], protect neurons from excitotoxic death [31], and may reduce cerebral edema and intracranial hypertension [47]. Susanto et al. demonstrated that compared to nonusers, either simvastatin 40 mg, atorvastatin 20 mg, or rosuvastatin 20 mg for ten days reduced mortality risk in TBI individuals. In contrast, statin discontinuation was associated with increased mortality [47].

Inflammatory Mediators and Cascades

Traumatic injuries cause disturbances in the normal cellular functioning of the brain due to the impact of direct, rotational, and shear forces [48]. Axonal injury leads to localized swelling, which hampers the transmission of signals. Traumatic injuries are also connected to alterations in cerebral blood flow, causing an initial decrease in blood circulation and subsequent unresponsive vasodilation, believed to be caused by the release of nitric oxide in the affected tissue [49].
Following a focal injury, the first component of the neuronal structure to be affected at the cellular level is the axonal membranes, given the rotational and direct forces. This axonal damage leads to the release of potassium from the intracellular environment. After membrane depolarization, calcium entry through voltage-dependent channels promotes the release of excitatory amino acids and neurotransmitters. At this point, potassium and calcium freely move between the intra- and extracellular spaces, disrupting intracellular homeostasis [48].
The impairment of intracellular functions leads the neuronal tissue into anaerobiosis [50]. Lactate levels rise, further contributing to local damage to the blood-brain barrier and cell death. This process can occur for 4 to 6 h [51].
In brief, the disruption of cellular membranes resulting from the primary mechanical insult or secondary injury triggers the release of damage-associated molecular patterns (DAMPs). This prompts the rapid upregulation of tumor necrosis factor (TNF), Interleukin 6 (IL-6), and Interleukin 1β (IL-1β) by local glial cells and infiltrating immune cells, acting as early mediators that drive the inflammatory response following traumatic injury [52].
In the context of TBI, microglia, play a crucial role. Traditionally, microglia have been classified into M1 (pro-inflammatory) and M2 (anti-inflammatory). This classification is based on their response to different micro-environmental disturbances 1, 2.
DAMPs release by injured neurons and proinflammatory and oxidative mediators from infiltrating immune cells leads to microglial cells polarization towards an M1-like phenotype [53]. The expression of proinflammatory factors (Table 1) such as IL-1β, TNF, IL-6, Nitric oxide synthase 2 (NOS2), Interleukin 12p40 (IL-12p40), and NADPH Oxidase 2 (NOX2) characterizes M1-like cells.
In response to anti-inflammatory and neurotrophic signals, microglia and macrophages can shift towards an M2-like phenotype [70]. M2-like cells express proteins such as CD206, CD163, arginase-1, FCγR, Ym1, IL-10, and TGFβ. Molecular pathways involved in regulating M2-like phenotypic transitions include STAT6/3, IRF-4/7, NF-κB p50/p50, Nrf2, and miR-124. M2-like microglia and macrophages release anti-inflammatory and trophic factors, promoting the resolution of inflammation [71,72]. Microglia and macrophages possess remarkable plasticity and can transition between M1-like and M2-like phenotypes. Mixed phenotypes are present in the acute phase following TBI, eventually transitioning to an M1-like dominant phenotype in the phase [72,73].
However, this binary classification has been interpreted somewhat arbitrarily, as there is a continuum of different intermediate phenotypes between M1 and M2, and microglia can transition from one phenotype to another, contributing to neurodegeneration, as seen in Alzheimer’s Disease animal models [72].
More recent studies have proposed a different categorization of microglia as resting, activated, or “disease-associated microglia” (DAM). DAM was initially characterized in mouse models of Alzheimer’s disease [71]. In DAM, homeostatic microglial markers are downregulated, and other genes, including Trem2, Apoe, Itgax, Clec7a, Axl, and Lpl, among others, are upregulated, playing a relevant job in the classification of microglia state. In this context, it has been proposed that rather than an initial M1 phenotype, in secondary TBI injury, the pathway to neurodegeneration follows an initial, activated state, and a progression DAM [71,72,73].
1.
Treatments based on the pathway level
The development of innovative anti-inflammatory drugs for managing TBI is facilitated by targeting various signaling pathways such as NF-κB, MAPKs, JAK/STAT, PI3K/Akt/mTOR, and TGF-β1 [74]. Once inflammatory mediators are released, immune response and glial cells are recruited. Microglial cells form the first line of differentiation between the intact and injured tissue [73,75]. Microglial cells release oxidation metabolites and pro-inflammatory reactants and cytokines such as interferon-gamma, interleukins, and tumor necrosis factor-alpha, especially in the latter. All of this leads to the stimulation of astrocytes for the formation of glial scars at the sites of trauma [27].
Nuclear factor-kappa B (NF-κB) is a transcription factor that regulates the synthesis of inflammatory molecules, pro-inflammatory cytokines, and chemokines [76]. In glial cells, NF-κB promotes inflammation, while in neurons, it is associated with neuroplasticity and neuronal development. Inhibiting this factor could reduce apoptosis and secondary inflammation in TBI [77,78].
In TBI, the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway decreases its expression, leading to increased cell death. In a study, recombinant erythropoietin was administered to cortical cells of rats with prior TBI, resulting in an increase in JAK and STAT and a reduction in apoptosis [79].
The MAPK (Mitogen-Activated Protein Kinase) pathway plays a crucial role in cell differentiation, proliferation, and survival. The path consists of c-Jun NH (2)-terminal kinase (JNK), extracellular signal-regulated protein kinase (ERK), and p38 [80]. Several studies have indicated that activating the p38 and JNK pathways contributes to increased neuronal damage in spinal cord injury and cerebral ischemia cases, activating a complex cascade in the mitochondria of brain cells and leading to apoptosis [81].
The dual nature of inflammation has been illustrated in experimental models exploring the involvement of TNF and inducible nitric oxide synthase (iNOS) following TBI [82]. TNF has been associated with brain edema, BBB disruption, and leukocyte recruitment [83]. Surprisingly, mice lacking TNF exhibited motor function impairment and larger lesions four weeks after the injury despite showing early neuroprotection [84]. Similarly, while TBI increased iNOS expression in the brain, which has multiple proinflammatory and neurotoxic effects, the genetic or chemical blockade of iNOS worsened spatial memory two to three weeks after the injury [85,86,87]. Cell death through programmed necrosis, such as necroptosis triggered by TNF-mediated receptor-interacting protein (RIP) kinase activation, can initiate a detrimental cycle: necrosis leads to further membrane disruption, promoting the release of DAMPs, which, in turn, exacerbates necrosis and amplifies inflammation [85,86,87].
2.
Other neuroinflammatory components
In response to TBI and glutamate toxicity, endogenous neuroprotectant adenosine levels are produced due to adenosine triphosphate and mRNA breakdown [88,89]. Following the ATP production and the mRNA, the activation of adenosine receptor A1 after TBI has been found to have anti-excitotoxic and anti-inflammatory effects in mice. However, it is essential to note that systemic administration of adenosine to patients can lead to bradycardia and hypotension [88,90].
Other components, including free radicals, lipid peroxidation, and direct impact, trigger the release of inflammatory mediators such as cytokines, chemokines, and complement. Cytokines are signaling molecules produced by immune system cells and brain cells, including microglia, astrocytes, and neurons. They act synergistically in cascades. After TBI, tumor necrosis factor α and interleukin1 β trigger the formation of peptides, ROS, and nitrogen species [91]. These mediators perpetuate secondary brain injury by activating arachidonic acid and coagulation cascades and disrupting BBB [92].
Blocking cytokines ameliorates pathological consequences in TBI models [93,94]. However, some cytokines modulate trophic responses and restorative functions [95] (Table 1). They can be divided into pro- and anti-inflammatory molecules.
3.
Specific treatment
The activation of inflammasomes, which leads to the release of IL-1β, can be inhibited by targeting IL-1 receptors, which can inhibit the invasion of circulating immune cells into the CNS [96].
Treatments like intravenous immunoglobulin can inhibit the priming of T cells from entering the CNS [97]. Furthermore, alterations in the gut microbiome may influence the balance between proinflammatory and anti-inflammatory T lymphocytes [98]. Impaired glymphatic clearance after TBI may hinder the removal of proinflammatory mediators, and ongoing investigations aim to enhance glymphatic flow, with increased clearance observed during sleep [99].
Nonsteroidal anti-inflammatory drugs (NSAIDs) produce an inhibition of COX that significantly reduces the levels of IL-1β and hinders the synthesis of IL-6 by modulating the pathways involved in producing vasodilator prostaglandins [100]. Another critical aspect of NSAIDs is their ability to stimulate the proliferator-activated receptor (PPAR), which elicits transcriptional regulatory effects that reduce the levels of various proinflammatory substances and decrease microglial activity. Additionally, several NSAIDs exhibit antioxidant properties and can inhibit the activation of NF-kB [101].

Mitochondrial Dysfunction

Impaired mitochondrial function and the generation of reactive oxygen species (ROS) are recognized consequences of direct and indirect traumatic brain injuries (TBIs) that contribute to the initiation of neuroinflammation [102]. Experimental evidence has shown that the movement of the phospholipid cardiolipin from the inner to the outer mitochondrial membrane occurs following TBI, marking damaged mitochondria for selective removal through mitophagy [103]. These mitochondrial signals of danger prompt local and systemic responses by interacting with specific receptors on immune cells [104]. For instance, mitochondrial DNA interacts with Toll-like receptor 9 (TLR9) on dendritic cells, while N-formyl peptides bind to formyl peptide receptor 1 on neutrophils [105].
After traumatic brain injury (TBI), energy storage may be depleted. Mitochondria play a crucial role in producing ATP. Neuronal cells maintain ATP levels through phosphocreatine deposits, glycolysis, and oxidative phosphorylation via the Krebs cycle and respiratory chain. The latter is the most efficient and occurs entirely in mitochondria. Lactate, a result of pyruvate oxidation, is the main substrate for energy production [105]. Impairments in the electron transport system (ETS) increase reactive oxygen species (ROS) formation and decrease energy production [106].
Mitochondrial dysfunction in severe head injury is supported by inadequate oxidative mitochondrial metabolism, mitochondrial swelling, and decreased ATP production [107,108]. Dysfunction begins early and may persist for days. Lipid peroxyl radicals contribute to secondary mitochondrial dysfunction [109]. Activation of the mitochondrial permeability transition pore (mPTP) determines cell survival after TBI [110]. Excess Ca2+ within mitochondria can cause mPTP opening, leading to mitochondrial edema and decreased oxidative metabolism [110]. mPTP opening allows extrusion of mitochondrial Ca2+ and activates harmful calcium-dependent proteases such as calpain [111]. Loss of cytochrome c through mPTP activates cellular apoptosis via caspase 9 [109,111]. Therapeutic interventions targeting mitochondria may delay or prevent secondary cascades leading to cell death and neurobehavioral disability [111].
  • Specific treatment
Cyclosporine A (CsA) stabilizes the mitochondrial permeability transition pore (mPTP) and has demonstrated neuroprotection in pre-clinical TBI studies [112]. Sullivan et al. showed on animal models that animals receiving CsA demonstrated a reduction in the lesion volume, even up to 74% [113]. Another mechanism that has been explored is the reduction of brain metabolic activity through hypothermia [113,114]. However, a recent meta-analysis conducted by Chen et al., which included 23 trials involving 2796 patients, demonstrated that therapeutic hypothermia did not reduce, but surprisingly, can increase the mortality rate of patients with TBI in some high-quality studies. However, the therapy can benefit patients with demonstrated increased intracranial pressure, not as a prophylactic therapy but as a therapy within the first 24 h [114].
Finally, barbiturates suppress cerebral metabolism and reduce cerebral blood volume and intracranial pressure (ICP) [115]. In a recent multicenter European trial, high-dose barbiturate treatment caused a decrease of 69% in ICP; however, this effect was also accompanied by hemodynamic instability, leading to more extended periods of mean arterial pressure <70 mmHg despite the use of vasopressors [115].

Cell Death

Cell death after TBI can occur through necrosis or apoptosis. Both can occur in regions remote from the impact site within days and weeks after trauma. Necrosis occurs in response to severe mechanical or ischemic/hypoxic tissue damage, excessive excitatory amino acid neurotransmitter release, and metabolic failure [116]. Caspases and calpain are important mediators of programmed cell death (apoptosis), with calpain activation more associated with necrosis [116].
Caspases are activated through the extrinsic pathway initiated by cell surface death receptor ligation (receptor-linked caspase-8 pathway) and the intrinsic pathway arising from mitochondria (mitochondrial caspase-9 pathway) [116]. Caspase 3 compromises membrane permeability to Ca2+, leading to elevated intracellular Ca2+ levels. Caspase 3 also degrades calpastatin, facilitating calpain activation [117]. Endogenous inhibitors, such as the inhibitors of the apoptosis family, modulate caspase activity within these pathways [117].
The activation of caspase-3 occurred via extrinsic and intrinsic apoptotic pathways after TBI has been documented, and this response is also mirrored in the retina [118]. However, experimental studies suggest that when activation is sustained, calpain is more important than caspase-3 in mediating cell death after TBI [118,119]. The anti-apoptotic modulator B-cell lymphoma (Bcl-2) inhibits mPTP, preserving mitochondrial homeostasis and preventing mitochondrial Ca2+ leak and programmed cell death [120]. Several in vivo studies with transgenic mice have shown promising results, at least with a reduced deficit in mice overexpressing the BCL-2 [121]. In clinical trials, a literature review published by Deng et al. showed reduced mortality and better outcomes in the Glasgow Coma Score (GOS) in the patients with increased Bcl-2 in the peritraumatic tissue, highlighting the importance of the peptide as a potential biomarker and therapeutic target [122].
  • Specific treatment
Erythropoietin (Epo) and its receptor (Epor) are expressed throughout the central nervous system [123]. Epo modulates caspase 1, caspase 3, and caspase 8-like activities, maintaining genomic DNA integrity and preventing acute cellular injury and microglial activation [124]. Epo also modulates mitochondrial membrane permeability and cytochrome c release, reduces cerebral vasospasm, and improves cerebral blood flow when given early post-injury [124].

3.3.2. BBB Disruption and Neutrophil Invasion

BBB regulates the exchange of substances between plasma and brain interstitium [125]. This barrier is formed by brain endothelial cells connected by tight junctions, with mechanical support from astrocytes critical for normal function [125]. Ion homeostasis and uptake of small molecules are conducted via specific endothelial membrane channels and solute carriers. Larger peptides and proteins are transported by endo- or transcytosis pathways within caveolae and clathrin-coated microvesicles. Paracellular diffusion is restricted by tight junctions between adjacent endothelial cells [126]. BBB disruption after injury is typically biphasic, with an immediate phase of hyperpermeability, temporary restoration of BBB function, and a delayed opening period [126]. BBB disruption leads to excess interstitial water (vasogenic edema) accumulation, contributing to cerebral swelling, brain shift, and herniation [125].
As previously mentioned, the BBB tight junction is based on the interaction between endothelial cells and astrocytes. Some studies have shown that the mechanical damage to astrocytes initiates oxidative-stress-mediated edema, altering astrocyte ionic gradients and increasing BBB permeability [125,127]. ROS increase brain endothelium permeability and promote post-traumatic invasion of inflammatory cells by upregulating endothelial expression of cell adhesion molecules such as intercellular adhesion molecule-1 (ICAM1) [127]. Degradation of membrane proteins and increased permeability contribute to BBB opening, vasogenic edema formation, and increased intracranial pressure [128].
Inflammatory cells provide the primary source of matrix metalloproteinase (MMP) activity [129]. MMPs promote cell death, including apoptosis [129]. Other inflammatory mediators in BBB opening include substance P, kinins, and bradykinins [128]. Particularly, substance P and its receptor, neurokinin 1 (NK1R), have been promising [130]. MMPs are released in ischemic brain injury and contribute to BBB disruption by degrading basal lamina components and tight junctions [129]. Later, MMPs are involved in tissue remodeling and neurovascular recovery. Vascular endothelial growth factor (VEGF) alters BBB permeability by changing the distribution and downregulating the expression of tight junction proteins [129].

Specific Treatment

Substance P antagonists (SP, NK1 receptor antagonist) show promising results in limiting BBB opening and edema after TBI [130]. Vink et al., in their review article, showed that NK1 antagonists can reduce posttraumatic ICP and improve brain oxygenation after TBI [90]. In vitro studies have shown the potential for the NK1-R antagonist to reverse the compromise, integrity, and function of the BBB [131].
Another alternative is Hyperbaric Oxygen Therapy (HBOT). HBOT reduces MMP-9 expression and inhibits neutrophilic infiltration. It may also counter capillary vasodilation within hypoxic tissues [132]. Hadanny et al. showed in their review that a search from 1969 to 2023 showed that HBOT should be recommended in acute moderate-severe TBI, specifically for patients suffering from prolonged post-concussion syndrome who have precise evidence of metabolic dysfunctional brain regions. However, further studies are needed to evaluate outcomes and determine the optimal treatment protocols [132] (Table 2).

4. Conclusions

Managing TBI patients includes specialized prehospital care, intensive clinical care, and long-term rehabilitation. However, neuroprotective agents to limit secondary injury or enhance repair lack clinical effectiveness. The complexity of TBI pathophysiology may reflect the difficulty of translating preclinical benefits into clinical practice. The current goal is to follow and identify the sequence of events of secondary lesions to avoid further neuronal damage. Knowledge of these concepts, the development of more efficient clinical trial designs, and the possibility of combination therapies may change the course of treatment in the acute phase in the near future.

Author Contributions

Conceptualization, R.L.O.A.; methodology, R.L.O.A., L.d.M.F., L.F.F., M.C.P.-C. and G.A.V.-G.; data curation, R.L.O.A., L.d.M.F. and L.F.F.; writing—original draft preparation, R.L.O.A., L.d.M.F., L.F.F., M.C.P.-C. and G.A.V.-G.; writing—review and editing, R.L.O.A., L.d.M.F., L.F.F., M.A., H.M., J.K.N., M.C.P.-C., G.A.V.-G. and G.J.H.; supervision, R.L.O.A.; project administration, R.L.O.A. 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

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Langlois, J.A.; Rutland-Brown, W.; Wald, M.M. The Epidemiology and Impact of Traumatic Brain Injury: A brief overview. J. Head Trauma Rehabil. 2006, 21, 375–378. [Google Scholar] [CrossRef]
  2. Ng, S.Y.; Lee, A.Y.W. Traumatic Brain Injuries: Pathophysiology and Potential Therapeutic Targets. Front. Cell. Neurosci. 2019, 13, 528. [Google Scholar] [CrossRef]
  3. Kaur, P.; Sharma, S. Recent Advances in Pathophysiology of Traumatic Brain Injury. Curr. Neuropharmacol. 2018, 16, 1224–1238. [Google Scholar] [CrossRef]
  4. Helmy, A.; Carpenter, K.L.; Menon, D.K.; Pickard, J.D.; Hutchinson, P.J. The Cytokine Response to Human Traumatic Brain Injury: Temporal Profiles and Evidence for Cerebral Parenchymal Production. J. Cereb. Blood Flow Metab. 2011, 31, 658–670. [Google Scholar] [CrossRef]
  5. Fakhry, S.M.; Trask, A.L.; Waller, M.A.; Watts, D.D. Management of Brain-Injured Patients by an Evidence-Based Medicine Protocol Improves Outcomes and Decreases Hospital Charges. J. Trauma Inj. Infect. Crit. Care 2004, 56, 492–500. [Google Scholar] [CrossRef]
  6. 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]
  7. Romeu-Mejia, R.; Giza, C.C.; Goldman, J.T. Concussion Pathophysiology and Injury Biomechanics. Curr. Rev. Musculoskelet. Med. 2019, 12, 105–116. [Google Scholar] [CrossRef]
  8. Girgis, F.; Pace, J.; Sweet, J.; Miller, J.P. Hippocampal Neurophysiologic Changes after Mild Traumatic Brain Injury and Potential Neuromodulation Treatment Approaches. Front. Syst. Neurosci. 2016, 10, 8. [Google Scholar] [CrossRef] [PubMed]
  9. Maas, A.I.R.; Stocchetti, N.; Bullock, R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008, 7, 728–741. [Google Scholar] [CrossRef] [PubMed]
  10. 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]
  11. Aromatario, M.; Torsello, A.; D’errico, S.; Bertozzi, G.; Sessa, F.; Cipolloni, L.; Baldari, B. Traumatic Epidural and Subdural Hematoma: Epidemiology, Outcome, and Dating. Medicina 2021, 57, 125. [Google Scholar] [CrossRef]
  12. Shin, D.-S.; Hwang, S.-C. Neurocritical Management of Traumatic Acute Subdural Hematomas. Korean J. Neurotrauma 2020, 16, 113–125. [Google Scholar] [CrossRef] [PubMed]
  13. Griswold, D.P.; Fernandez, L.L.; Rubiano, A.M. Traumatic Subarachnoid Hemorrhage: A Scoping Review. J. Neurotrauma 2022, 39, 35–48. [Google Scholar] [CrossRef] [PubMed]
  14. Vinciguerra, L.; Bösel, J. Noninvasive Neuromonitoring: Current Utility in Subarachnoid Hemorrhage, Traumatic Brain Injury, and Stroke. Neurocritical Care 2017, 27, 122–140. [Google Scholar] [CrossRef] [PubMed]
  15. Adatia, K.; Newcombe, V.F.J.; Menon, D.K. Contusion Progression Following Traumatic Brain Injury: A Review of Clinical and Radiological Predictors, and Influence on Outcome. Neurocritical Care 2021, 34, 312–324. [Google Scholar] [CrossRef] [PubMed]
  16. Sturiale, C.L.; De Bonis, P.; Rigante, L.; Calandrelli, R.; D’Arrigo, S.; Pompucci, A.; Mangiola, A.; D’Apolito, G.; Colosimo, C.; Anile, C. Do traumatic brain contusions increase in size after decompressive craniectomy? J. Neurotrauma 2012, 29, 2723–2726. [Google Scholar] [CrossRef] [PubMed]
  17. Simard, J.M.; Kilbourne, M.; Tsymbalyuk, O.; Tosun, C.; Caridi, J.; Ivanova, S.; Keledjian, K.; Bochicchio, G.; Gerzanich, V. Key role of sulfonylurea receptor 1 in progressive secondary hemorrhage after brain contusion. J. Neurotrauma 2009, 26, 2257–2267. [Google Scholar] [CrossRef]
  18. Kurland, D.; Hong, C.; Aarabi, B.; Gerzanich, V.; Simard, J.M. Hemorrhagic progression of a contusion after traumatic brain injury: A review. J. Neurotrauma 2012, 29, 19–31. [Google Scholar] [CrossRef]
  19. Smith, D.H.; Meaney, D.F.; Shull, W.H. Diffuse Axonal Injury in Head Trauma. J. Head Trauma Rehabil. 2003, 18, 307–316. [Google Scholar] [CrossRef]
  20. Humble, S.S.; Wilson, L.D.; Wang, L.; Long, D.A.; Smith, M.A.; Siktberg, J.C.; Mirhoseini, M.F.; Bhatia, A.; Pruthi, S.; Day, M.A.; et al. Prognosis of diffuse axonal injury with traumatic brain injury. J. Trauma Inj. Infect. Crit. Care Surg. 2018, 85, 155–159. [Google Scholar] [CrossRef]
  21. Johnson, V.E.; Stewart, W.; Smith, D.H. Axonal pathology in traumatic brain injury. Exp. Neurol. 2013, 246, 35–43. [Google Scholar] [CrossRef]
  22. Takahashi, C.E.; Virmani, D.; Chung, D.Y.; Ong, C.; Cervantes-Arslanian, A.M. Blunt and Penetrating Severe Traumatic Brain Injury. Neurol. Clin. 2021, 39, 443–469. [Google Scholar] [CrossRef]
  23. Fathalla, H.; Ashry, A.; El-Fiki, A. Managing military penetrating brain injuries in the war zone: Lessons learned. Neurosurg. Focus 2018, 45, E6. [Google Scholar] [CrossRef]
  24. Solumsmoen, S.; Lilja-Cyron, A.; Buch, K.F.; Kelsen, J. Traumatic penetrating brain injury. Ugeskr Laeger 2018, 180, V03180201. [Google Scholar]
  25. Raghupathi, R. Cell Death Mechanisms Following Traumatic Brain Injury. Brain Pathol. 2004, 14, 215–222. [Google Scholar] [CrossRef]
  26. Kahriman, A.; Bouley, J.; Smith, T.W.; Bosco, D.A.; Woerman, A.L.; Henninger, N. Mouse closed head traumatic brain injury replicates the histological tau pathology pattern of human disease: Characterization of a novel model and systematic review of the literature. Acta Neuropathol. Commun. 2021, 9, 118. [Google Scholar] [CrossRef] [PubMed]
  27. Zhao, L.; Wang, W.; Zhong, J.; Li, Y.; Cheng, Y.; Su, Z.; Zheng, W.; Guan, X.-D. The effects of magnesium sulfate therapy after severe diffuse axonal injury. Ther. Clin. Risk Manag. 2016, 12, 1481–1486. [Google Scholar] [CrossRef] [PubMed]
  28. Lauritzen, M.; Dreier, J.P.; Fabricius, M.; Hartings, J.A.; Graf, R.; Strong, A.J. Clinical Relevance of Cortical Spreading Depression in Neurological Disorders: Migraine, Malignant Stroke, Subarachnoid and Intracranial Hemorrhage, and Traumatic Brain Injury. J. Cereb. Blood Flow Metab. 2011, 31, 17–35. [Google Scholar] [CrossRef]
  29. Timofeev, I.; Carpenter, K.L.H.; Nortje, J.; Al-Rawi, P.G.; O’Connell, M.T.; Czosnyka, M.; Smielewski, P.; Pickard, J.D.; Menon, D.K.; Kirkpatrick, P.J.; et al. Cerebral extracellular chemistry and outcome following traumatic brain injury: A microdialysis study of 223 patients. Brain 2011, 134, 484–494. [Google Scholar] [CrossRef]
  30. Verweij, B.H.; Muizelaar, J.P.; Vinas, F.C.; Peterson, P.L.; Xiong, Y.; Lee, C.P. Impaired cerebral mitochondrial function after traumatic brain injury in humans. J. Neurosurg. 2000, 93, 815–820. [Google Scholar] [CrossRef] [PubMed]
  31. Wible, E.F.; Laskowitz, D.T. Statins in Traumatic Brain Injury. Neurotherapeutics 2010, 7, 62–73. [Google Scholar] [CrossRef] [PubMed]
  32. Baracaldo-Santamaría, D.; Ariza-Salamanca, D.F.; Corrales-Hernández, M.G.; Pachón-Londoño, M.J.; Hernandez-Duarte, I.; Calderon-Ospina, C.-A. Revisiting Excitotoxicity in Traumatic Brain Injury: From Bench to Bedside. Pharmaceutics 2022, 14, 152. [Google Scholar] [CrossRef]
  33. Ikonomidou, C.; Turski, L. Why did NMDA receptor antagonists fail clinical trials for stroke and traumatic brain injury? Lancet Neurol. 2002, 1, 383–386. [Google Scholar] [CrossRef]
  34. Shohami, E.; Biegon, A. Novel Approach to the Role of NMDA Receptors in Traumatic Brain Injury. CNS Neurol. Disord.-Drug Targets 2014, 13, 567–573. [Google Scholar] [CrossRef]
  35. DI Vergouwen, M.; 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]
  36. Li, W.; Bai, Y.-A.; Li, Y.-J.; Liu, K.-G.; Wang, M.-D.; Xu, G.-Z.; Shang, H.-L.; Li, Y.-F. Magnesium Sulfate for Acute Traumatic Brain Injury. J. Craniofacial Surg. 2015, 26, 393–398. [Google Scholar] [CrossRef] [PubMed]
  37. Temkin, N.R.; Anderson, G.D.; Winn, H.R.; Ellenbogen, R.G.; Britz, G.W.; Schuster, J.; Lucas, T.; Newell, D.W.; Mansfield, P.N.; Machamer, J.E.; et al. Magnesium sulfate for neuroprotection after traumatic brain injury: A randomised controlled trial. Lancet Neurol. 2007, 6, 29–38. [Google Scholar] [CrossRef] [PubMed]
  38. Temkin, N.; Machamer, J.; Dikmen, S.; Nelson, L.D.; Barber, J.; Hwang, P.H.; Boase, K.; Stein, M.B.; Sun, X.; Giacino, J.; et al. Risk Factors for High Symptom Burden Three Months after Traumatic Brain Injury and Implications for Clinical Trial Design: A Transforming Research and Clinical Knowledge in Traumatic Brain Injury Study. J. Neurotrauma 2022, 39, 1524–1532. [Google Scholar] [CrossRef]
  39. Xiong, Y.; Mahmood, A.; Chopp, M. Animal models of traumatic brain injury. Nat. Rev. Neurosci. 2013, 14, 128–142. [Google Scholar] [CrossRef]
  40. Zhang, Z.-D.; Fang, H.-Y.; Pang, C.; Yang, Y.; Li, S.-Z.; Zhou, L.-L.; Bai, G.-H.; Sheng, H.-S. Giant Pediatric Supratentorial Tumor: Clinical Feature and Surgical Strategy. Front. Pediatr. 2022, 10, 870951. [Google Scholar] [CrossRef]
  41. Besson, V.C. Drug targets for traumatic brain injury from poly(ADP-ribose)polymerase pathway modulation. Br. J. Pharmacol. 2009, 157, 695–704. [Google Scholar] [CrossRef]
  42. O’connell, K.M.; Littleton-Kearney, M.T. The Role of Free Radicals in Traumatic Brain Injury. Biol. Res. Nurs. 2013, 15, 253–263. [Google Scholar] [CrossRef]
  43. Darwish, R.S.; Amiridze, N.; Aarabi, B. Nitrotyrosine as an Oxidative Stress Marker: Evidence for Involvement in Neurologic Outcome in Human Traumatic Brain Injury. J. Trauma Inj. Infect. Crit. Care 2007, 63, 439–442. [Google Scholar] [CrossRef]
  44. Nisenbaum, E.J.; Novikov, D.S.; Lui, Y.W. The Presence and Role of Iron in Mild Traumatic Brain Injury: An Imaging Perspective. J. Neurotrauma 2014, 31, 301–307. [Google Scholar] [CrossRef]
  45. Secades, J.J. Role of Citicoline in the Management of Traumatic Brain Injury. Pharmaceuticals 2021, 14, 410. [Google Scholar] [CrossRef]
  46. Zafonte, R.D.; Bagiella, E.; Ansel, B.M.; Novack, T.A.; Friedewald, W.T.; Hesdorffer, D.C.; Timmons, S.D.; Jallo, J.; Eisenberg, H.; Hart, T.; et al. Effect of Citicoline on Functional and Cognitive Status Among Patients with Traumatic Brain Injury: Citicoline Brain Injury Treatment Trial (COBRIT). JAMA 2012, 308, 1993–2000. [Google Scholar] [CrossRef]
  47. Susanto, M.; Siahaan, A.M.P.; Wirjomartani, B.A.; Setiawan, H.; Aryanti, C. Michael The neuroprotective effect of statin in traumatic brain injury: A systematic review. World Neurosurg. X 2023, 19, 100211. [Google Scholar] [CrossRef]
  48. Capizzi, A.; Woo, J.; Verduzco-Gutierrez, M. Traumatic Brain Injury. Med. Clin. N. Am. 2020, 104, 213–238. [Google Scholar] [CrossRef] [PubMed]
  49. Greco, T.; Vespa, P.M.; Prins, M.L. Alternative substrate metabolism depends on cerebral metabolic state following traumatic brain injury. Exp. Neurol. 2020, 329, 113289. [Google Scholar] [CrossRef] [PubMed]
  50. Dixon, K.J. Pathophysiology of Traumatic Brain Injury. Phys. Med. Rehabil. Clin. N. Am. 2017, 28, 215–225. [Google Scholar] [CrossRef] [PubMed]
  51. Block, M.L.; Hong, J.-S. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Prog. Neurobiol. 2005, 76, 77–98. [Google Scholar] [CrossRef] [PubMed]
  52. Corps, K.N.; Roth, T.L.; McGavern, D.B. Inflammation and Neuroprotection in Traumatic Brain Injury. JAMA Neurol. 2015, 72, 355–362. [Google Scholar] [CrossRef] [PubMed]
  53. Guilfoyle, M.R.; Helmy, A.; Donnelly, J.; Stovell, M.G.; Timofeev, I.; Pickard, J.D.; Czosnyka, M.; Smielewski, P.; Menon, D.K.; Carpenter, K.L.H.; et al. Characterising the dynamics of cerebral metabolic dysfunction following traumatic brain injury: A microdialysis study in 619 patients. PLoS ONE 2021, 16, e0260291. [Google Scholar] [CrossRef] [PubMed]
  54. Helmy, A.; Guilfoyle, M.R.; Carpenter, K.L.; Pickard, J.D.; Menon, D.K.; Hutchinson, P.J. Recombinant human interleukin-1 receptor antagonist in severe traumatic brain injury: A phase II randomized control trial. J. Cereb. Blood Flow Metab. 2014, 34, 845–851. [Google Scholar] [CrossRef] [PubMed]
  55. Santacruz, C.A.; Vincent, J.-L.; Bader, A.; Rincón-Gutiérrez, L.A.; Dominguez-Curell, C.; Communi, D.; Taccone, F.S. Association of cerebrospinal fluid protein biomarkers with outcomes in patients with traumatic and non-traumatic acute brain injury: Systematic review of the literature. Crit. Care 2021, 25, 278. [Google Scholar] [CrossRef] [PubMed]
  56. Longhi, L.; Perego, C.; Ortolano, F.; Aresi, S.; Fumagalli, S.; Zanier, E.R.; Stocchetti, N.; De Simoni, M.-G. Tumor necrosis factor in traumatic brain injury: Effects of genetic deletion of p55 or p75 receptor. J. Cereb. Blood Flow Metab. 2013, 33, 1182–1189. [Google Scholar] [CrossRef] [PubMed]
  57. Rowe, R.K.; Harrison, J.L.; Zhang, H.; Bachstetter, A.D.; Hesson, D.P.; O’hara, B.F.; Greene, M.I.; Lifshitz, J. Novel TNF receptor-1 inhibitors identified as potential therapeutic candidates for traumatic brain injury. J. Neuroinflamm. 2018, 15, 154. [Google Scholar] [CrossRef] [PubMed]
  58. Shohami, E.; Ginis, I.; Hallenbeck, J.M. Dual role of tumor necrosis factor alpha in brain injury. Cytokine Growth Factor Rev. 1999, 10, 119–130. [Google Scholar] [CrossRef]
  59. Ooi, S.Z.Y.; Spencer, R.J.; Hodgson, M.; Mehta, S.; Phillips, N.L.; Preest, G.; Manivannan, S.; Wise, M.P.; Galea, J.; Zaben, M. Interleukin-6 as a prognostic biomarker of clinical outcomes after traumatic brain injury: A systematic review. Neurosurg. Rev. 2022, 45, 3035–3054. [Google Scholar] [CrossRef]
  60. Tsitsipanis, C.; Miliaraki, M.; Paflioti, E.; Lazarioti, S.; Moustakis, N.; Ntotsikas, K.; Theofanopoulos, A.; Ilia, S.; Vakis, A.; Simos, P.; et al. Inflammation biomarkers IL-6 and IL-10 may improve the diagnostic and prognostic accuracy of currently authorized traumatic brain injury tools. Exp. Ther. Med. 2023, 26, 364. [Google Scholar] [CrossRef]
  61. Hammad, A.; Westacott, L.; Zaben, M. The role of the complement system in traumatic brain injury: A review. J. Neuroinflamm. 2018, 15, 24. [Google Scholar] [CrossRef]
  62. Mallah, K.; Couch, C.; Alshareef, M.; Borucki, D.; Yang, X.; Alawieh, A.; Tomlinson, S. Complement mediates neuroinflammation and cognitive decline at extended chronic time points after traumatic brain injury. Acta Neuropathol. Commun. 2021, 9, 72. [Google Scholar] [CrossRef]
  63. van Erp, I.A.M.; Michailidou, I.; van Essen, T.A.; van der Jagt, M.; Moojen, W.; Peul, W.C.; Baas, F.; Fluiter, K. Tackling Neuroinflammation after Traumatic Brain Injury: Complement Inhibition as a Therapy for Secondary Injury. Neurotherapeutics 2022, 20, 284–303. [Google Scholar] [CrossRef]
  64. Angiopoietin-1 Reduces Vascular Endothelial Growth Factor-Induced Brain Endothelial Permeability via Upregulation of ZO-2-PubMed. (n.d.). Available online: https://pubmed.ncbi.nlm.nih.gov/19148554/ (accessed on 27 December 2023).
  65. Brickler, T.R.; Hazy, A.; Correa, F.G.; Dai, R.; Kowalski, E.J.; Dickerson, R.; Chen, J.; Wang, X.; Morton, P.D.; Whittington, A.; et al. Angiopoietin/Tie2 Axis Regulates the Age-at-Injury Cerebrovascular Response to Traumatic Brain Injury. J. Neurosci. 2018, 38, 9618–9634. [Google Scholar] [CrossRef]
  66. Kim, H.; Lee, J.M.; Park, J.S.; Jo, S.A.; Kim, Y.-O.; Kim, C.-W.; Jo, I. Dexamethasone coordinately regulates angiopoietin-1 and VEGF: A mechanism of glucocorticoid-induced stabilization of blood–brain barrier. Biochem. Biophys. Res. Commun. 2008, 372, 243–248. [Google Scholar] [CrossRef] [PubMed]
  67. Michinaga, S.; Tanabe, A.; Nakaya, R.; Fukutome, C.; Inoue, A.; Iwane, A.; Minato, Y.; Tujiuchi, Y.; Miyake, D.; Mizuguchi, H.; et al. Angiopoietin-1/Tie-2 signal after focal traumatic brain injury is potentiated by BQ788, an ETB receptor antagonist, in the mouse cerebrum: Involvement in recovery of blood–brain barrier function. J. Neurochem. 2020, 154, 330–348. [Google Scholar] [CrossRef] [PubMed]
  68. Nag, S.; Papneja, T.; Venugopalan, R.; Stewart, D.J. Increased angiopoietin2 expression is associated with endothelial apoptosis and blood–brain barrier breakdown. Lab. Investig. J. Tech. Methods Pathol. 2005, 85, 1189–1198. [Google Scholar] [CrossRef] [PubMed]
  69. Morganti-Kossmann, M.C.; Rancan, M.; Stahel, P.F.; Kossmann, T. Inflammatory response in acute traumatic brain injury: A double-edged sword. Curr. Opin. Crit. Care 2002, 8, 101–105. [Google Scholar] [CrossRef] [PubMed]
  70. David, S.; Kroner, A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat. Rev. Neurosci. 2011, 12, 388–399. [Google Scholar] [CrossRef]
  71. Kumar, A.; Loane, D.J. Neuroinflammation after traumatic brain injury: Opportunities for therapeutic intervention. Brain Behav. Immun. 2012, 26, 1191–1201. [Google Scholar] [CrossRef] [PubMed]
  72. Loane, D.J.; Kumar, A.; Stoica, B.A.; Cabatbat, R.; Faden, A.I. Progressive Neurodegeneration After Experimental Brain Trauma: Association with Microglial Activation. J. Neuropathol. Exp. Neurol. 2014, 73, 14–29. [Google Scholar] [CrossRef]
  73. Kalra, S.; Malik, R.; Singh, G.; Bhatia, S.; Al-Harrasi, A.; Mohan, S.; Albratty, M.; Albarrati, A.; Tambuwala, M.M. Pathogenesis and management of traumatic brain injury (TBI): Role of neuroinflammation and anti-inflammatory drugs. Inflammopharmacology 2022, 30, 1153–1166. [Google Scholar] [CrossRef] [PubMed]
  74. Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.-B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752–758. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, A.P.-Y.; Tung, J.Y.-L.; Ku, D.T.-L.; Luk, C.-W.; Ling, A.S.-C.; Kwong, D.L.-W.; Cheng, K.K.-F.; Ho, W.W.-S.; Shing, M.M.-K.; Chan, G.C.-F. Outcome of Chinese children with craniopharyngioma: A 20-year population-based study by the Hong Kong Pediatric Hematology/Oncology Study Group. Child’s Nerv. Syst. 2020, 36, 497–505. [Google Scholar] [CrossRef] [PubMed]
  76. Mattson, M.P.; Camandola, S. NF-κB in neuronal plasticity and neurodegenerative disorders. J. Clin. Investig. 2001, 107, 247–254. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, K.; Mu, X.S.; Hayes, R.L. Increased cortical nuclear factor-κB (NF-κB) DNA binding activity after traumatic brain injury in rats. Neurosci. Lett. 1995, 197, 101–104. [Google Scholar] [CrossRef] [PubMed]
  78. Oliva, A.A., Jr.; Kang, Y.; Sanchez-Molano, J.; Furones, C.; Atkins, C.M. STAT3 signaling after traumatic brain injury: STAT3 Activation after Traumatic Brain Injury. J. Neurochem. 2012, 120, 710–720. [Google Scholar] [CrossRef]
  79. Carbonell, W.S.; Mandell, J.W. Transient Neuronal but Persistent Astroglial Activation of ERK/MAP Kinase after Focal Brain Injury in Mice. J. Neurotrauma 2003, 20, 327–336. [Google Scholar] [CrossRef] [PubMed]
  80. Dietrich, W.D.; Bramlett, H.M. Therapeutic hypothermia and targeted temperature management in traumatic brain injury: Clinical challenges for successful translation. Brain Res. 2016, 1640, 94–103. [Google Scholar] [CrossRef]
  81. Ziebell, J.M.; Morganti-Kossmann, M.C. Involvement of Pro- and Anti-Inflammatory Cytokines and Chemokines in the Pathophysiology of Traumatic Brain Injury. Neurotherapeutics 2010, 7, 22–30. [Google Scholar] [CrossRef]
  82. Scherbel, U.; Raghupathi, R.; Nakamura, M.; Saatman, K.E.; Trojanowski, J.Q.; Neugebauer, E.; Marino, M.W.; McIntosh, T.K. Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury. Proc. Natl. Acad. Sci. USA 1999, 96, 8721–8726. [Google Scholar] [CrossRef]
  83. Sinz, E.H.; Kochanek, P.M.; Dixon, C.E.; Clark, R.S.; Carcillo, J.A.; Schiding, J.K.; Chen, M.; Wisniewski, S.R.; Carlos, T.M.; Williams, D.; et al. Inducible nitric oxide synthase is an endogenous neuroprotectant after traumatic brain injury in rats and mice. J. Clin. Investig. 1999, 104, 647–656. [Google Scholar] [CrossRef]
  84. Au, A.K.; Aneja, R.K.; Bell, M.J.; Bayir, H.; Feldman, K.; Adelson, P.D.; Fink, E.L.; Kochanek, P.M.; Clark, R.S. Cerebrospinal Fluid Levels of High-Mobility Group Box 1 and Cytochrome C Predict Outcome after Pediatric Traumatic Brain Injury. J. Neurotrauma 2012, 29, 2013–2021. [Google Scholar] [CrossRef]
  85. Laird, M.D.; Shields, J.S.; Sukumari-Ramesh, S.; Kimbler, D.E.; Fessler, R.D.; Shakir, B.; Youssef, P.; Yanasak, N.; Vender, J.R.; Dhandapani, K.M. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia 2014, 62, 26–38. [Google Scholar] [CrossRef]
  86. You, Z.; Savitz, S.I.; Yang, J.; Degterev, A.; Yuan, J.; Cuny, G.D.; Moskowitz, M.A.; Whalen, M.J. Necrostatin-1 Reduces Histopathology and Improves Functional Outcome after Controlled Cortical Impact in Mice. J. Cereb. Blood Flow Metab. 2008, 28, 1564–1573. [Google Scholar] [CrossRef]
  87. Kochanek, P.M.; Clark, R.S.; Ruppel, R.A.; Adelson, P.D.; Bell, M.J.; Whalen, M.J.; Robertson, C.L.; Satchell, M.A.; Seidberg, N.A.; Marion, D.W.; et al. Biochemical, cellular, and molecular mechanisms in the evolution of secondary damage after severe traumatic brain injury in infants and children: Lessons learned from the bedside. Pediatr. Crit. Care Med. 2000, 1, 4–19. [Google Scholar] [CrossRef] [PubMed]
  88. Viviani, B.; Boraso, M.; Marchetti, N.; Marinovich, M. Perspectives on neuroinflammation and excitotoxicity: A neurotoxic conspiracy? NeuroToxicology 2014, 43, 10–20. [Google Scholar] [CrossRef] [PubMed]
  89. Verrier, J.D.; Exo, J.L.; Jackson, T.C.; Ren, J.; Gillespie, D.G.; Dubey, R.K.; Kochanek, P.M.; Jackson, E.K. Expression of the 2′,3′-cAMP-adenosine pathway in astrocytes and microglia: 2′,3′-cAMP Metabolism in Astrocytes and Microglia. J. Neurochem. 2011, 118, 979–987. [Google Scholar] [CrossRef] [PubMed]
  90. Cederberg, D.; Siesjö, P. What has inflammation to do with traumatic brain injury? Child’s Nerv. Syst. 2010, 26, 221–226. [Google Scholar] [CrossRef] [PubMed]
  91. Dohi, K.; Kraemer, B.C.; Erickson, M.; McMillan, P.J.; Kovac, A.; Flachbartova, Z.; Hansen, K.M.; Shah, G.N.; Sheibani, N.; Salameh, T.; et al. Molecular Hydrogen in Drinking Water Protects against Neurodegenerative Changes Induced by Traumatic Brain Injury. PLoS ONE 2014, 9, e108034. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, S.; Hsu, C.; Huang, W.; Wang, J. Post-injury baicalein improves histological and functional outcomes and reduces inflammatory cytokines after experimental traumatic brain injury: Baicalein Reduces Cytokine Expression in TBI. Br. J. Pharmacol. 2008, 155, 1279–1296. [Google Scholar] [CrossRef]
  93. Kokiko-Cochran, O.N.; Godbout, J.P. The Inflammatory Continuum of Traumatic Brain Injury and Alzheimer’s Disease. Front. Immunol. 2018, 9, 672. [Google Scholar] [CrossRef]
  94. Thelin, E.P.; Hall, C.E.; Gupta, K.; Carpenter, K.L.; Chandran, S.; Hutchinson, P.J.; Patani, R.; Helmy, A. Elucidating Pro-Inflammatory Cytokine Responses after Traumatic Brain Injury in a Human Stem Cell Model. J. Neurotrauma 2018, 35, 341–352. [Google Scholar] [CrossRef]
  95. Anderson, G.D.; Peterson, T.C.; Haar, C.V.; Kantor, E.D.; Farin, F.M.; Bammler, T.K.; MacDonald, J.W.; Hoane, M.R. Comparison of the effects of erythropoietin and anakinra on functional recovery and gene expression in a traumatic brain injury model. Front. Pharmacol. 2013, 4, 129. [Google Scholar] [CrossRef] [PubMed]
  96. Simon, D.W.; McGeachy, M.J.; Bayır, H.; Clark, R.S.B.; Loane, D.J.; Kochanek, P.M. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat. Rev. Neurol. 2017, 13, 171–191. [Google Scholar] [CrossRef] [PubMed]
  97. Mazzeo, A.T.; Kunene, N.K.; Gilman, C.B.; Hamm, R.J.; Hafez, N.; Bullock, M.R. Severe Human Traumatic Brain Injury, but Not Cyclosporin A Treatment, Depresses Activated T Lymphocytes Early after Injury. J. Neurotrauma 2006, 23, 962–975. [Google Scholar] [CrossRef] [PubMed]
  98. Jin, X.; Ishii, H.; Bai, Z.; Itokazu, T.; Yamashita, T. Temporal Changes in Cell Marker Expression and Cellular Infiltration in a Controlled Cortical Impact Model in Adult Male C57BL/6 Mice. PLoS ONE 2012, 7, e41892. [Google Scholar] [CrossRef]
  99. Breitner, J.C. Inflammatory processes and antiinflammatory drugs in Alzheimer’s disease: A current appraisal. Neurobiol. Aging 1996, 17, 789–794. [Google Scholar] [CrossRef]
  100. Grilli, M.; Pizzi, M.; Memo, M.; Spano, P. Neuroprotection by Aspirin and Sodium Salicylate Through Blockade of NF-κB Activation. Science 1996, 274, 1383–1385. [Google Scholar] [CrossRef]
  101. Suliman, H.B.; Piantadosi, C.A. Mitochondrial Quality Control as a Therapeutic Target. Pharmacol. Rev. 2016, 68, 20–48. [Google Scholar] [CrossRef]
  102. Chu, T.C.; Ji, J.; Dagda, R.K.; Jiang, J.F.; Tyurina, Y.Y.; Kapralov, A.A.; Tyurin, V.A.; Yanamala, N.; Shrivastava, I.H.; Mohammadyani, D.; et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat. Cell Biol. 2013, 15, 1197–1205. [Google Scholar] [CrossRef] [PubMed]
  103. Iyer, S.S.; He, Q.; Janczy, J.R.; Elliott, E.I.; Zhong, Z.; Olivier, A.K.; Sadler, J.J.; Knepper-Adrian, V.; Han, R.; Qiao, L.; et al. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity 2013, 39, 311–323. [Google Scholar] [CrossRef] [PubMed]
  104. Schurr, A.; Payne, R. Lactate, not pyruvate, is neuronal aerobic glycolysis end product: An in vitro electrophysiological study. Neuroscience 2007, 147, 613–619. [Google Scholar] [CrossRef] [PubMed]
  105. Zauner, A.; Doppenberg, E.; Woodward, J.; Allen, C.; Jebraili, S.; Young, H.; Bullock, R. Multiparametric continuous monitoring of brain metabolism and substrate delivery in neurosurgical patients. Neurol. Res. 1997, 19, 265–273. [Google Scholar] [CrossRef] [PubMed]
  106. Hiebert, J.B.; Shen, Q.; Thimmesch, A.R.; Pierce, J.D. Traumatic Brain Injury and Mitochondrial Dysfunction. Am. J. Med. Sci. 2015, 350, 132–138. [Google Scholar] [CrossRef] [PubMed]
  107. Cheng, G.; Kong, R.; Zhang, L.; Zhang, J. Mitochondria in traumatic brain injury and mitochondrial-targeted multipotential therapeutic strategies. Br. J. Pharmacol. 2012, 167, 699–719. [Google Scholar] [CrossRef]
  108. Ahluwalia, M.; Kumar, M.; Ahluwalia, P.; Rahimi, S.; Vender, J.R.; Raju, R.P.; Hess, D.C.; Baban, B.; Vale, F.L.; Dhandapani, K.M.; et al. Rescuing mitochondria in traumatic brain injury and intracerebral hemorrhages—A potential therapeutic approach. Neurochem. Int. 2021, 150, 105192. [Google Scholar] [CrossRef]
  109. McGovern, A.J.; Barreto, G.E. Mitochondria dysfunction and inflammation in traumatic brain injury: Androgens to the battlefront. Androg. Clin. Res. Ther. 2021, 2, 304–315. [Google Scholar] [CrossRef]
  110. Hill, R.L.; Kulbe, J.R.; Singh, I.N.; Wang, J.A.; Hall, E.D. Synaptic Mitochondria are More Susceptible to Traumatic Brain Injury-induced Oxidative Damage and Respiratory Dysfunction than Non-synaptic Mitochondria. Neuroscience 2018, 386, 265–283. [Google Scholar] [CrossRef]
  111. Li, J.-L.; Lin, T.-Y.; Chen, P.-L.; Guo, T.-N.; Huang, S.-Y.; Chen, C.-H.; Lin, C.-H.; Chan, C.-C. Mitochondrial Function and Parkinson’s Disease: From the Perspective of the Electron Transport Chain. Front. Mol. Neurosci. 2021, 14, 797833. [Google Scholar] [CrossRef] [PubMed]
  112. Mustafa, A.G.; Alshboul, O.A. Pathophysiology of traumatic brain injury. Neurosciences 2013, 18, 222–234. [Google Scholar] [PubMed]
  113. Sullivan, J.P.; Nahed, B.V.; Madden, M.W.; Oliveira, S.M.; Springer, S.; Bhere, D.; Chi, A.S.; Wakimoto, H.; Rothenberg, S.M.; Sequist, L.V.; et al. Brain Tumor Cells in Circulation Are Enriched for Mesenchymal Gene Expression. Cancer Discov. 2014, 4, 1299–1309. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, H.; Wu, F.; Yang, P.; Shao, J.; Chen, Q.; Zheng, R. A meta-analysis of the effects of therapeutic hypothermia in adult patients with traumatic brain injury. Crit. Care 2019, 23, 396. [Google Scholar] [CrossRef] [PubMed]
  115. Majdan, M.; Mauritz, W.; Wilbacher, I.; Brazinova, A.; Rusnak, M.; Leitgeb, J. Barbiturates Use and Its Effects in Patients with Severe Traumatic Brain Injury in Five European Countries. J. Neurotrauma 2013, 30, 23–29. [Google Scholar] [CrossRef] [PubMed]
  116. Werner, C.; Engelhard, K. Pathophysiology of traumatic brain injury. Br. J. Anaesth. 2007, 99, 4–9. [Google Scholar] [CrossRef]
  117. Eldadah, B.A.; Faden, A.I. Caspase Pathways, Neuronal Apoptosis, and CNS Injury. J. Neurotrauma 2000, 17, 811–829. [Google Scholar] [CrossRef] [PubMed]
  118. Kovács-Öller, T.; Zempléni, R.; Balogh, B.; Szarka, G.; Fazekas, B.; Tengölics, J.; Amrein, K.; Czeiter, E.; Hernádi, I.; Büki, A.; et al. Traumatic Brain Injury Induces Microglial and Caspase3 Activation in the Retina. Int. J. Mol. Sci. 2023, 24, 4451. [Google Scholar] [CrossRef] [PubMed]
  119. Saatman, K.E.; Creed, J.; Raghupathi, R. Calpain as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2010, 7, 31–42. [Google Scholar] [CrossRef]
  120. Taylor, R.C.; Cullen, S.P.; Martin, S.J. Apoptosis: Controlled demolition at the cellular level. Nat. Rev. Mol. Cell Biol. 2008, 9, 231–241. [Google Scholar] [CrossRef]
  121. Raghupathi, R.; Fernandez, S.C.; Murai, H.; Trusko, S.P.; Scott, R.W.; Nishioka, W.K.; McIntosh, T.K. BCL-2 Overexpression Attenuates Cortical Cell Loss after Traumatic Brain Injury in Transgenic Mice. J. Cereb. Blood Flow Metab. 1998, 18, 1259–1269. [Google Scholar] [CrossRef]
  122. Deng, H.; Yue, J.K.; Zusman, B.E.; Nwachuku, E.L.; Abou-Al-Shaar, H.; Upadhyayula, P.S.; Okonkwo, D.O.; Puccio, A.M. B-Cell Lymphoma 2 (Bcl-2) and Regulation of Apoptosis after Traumatic Brain Injury: A Clinical Perspective. Medicina 2020, 56, 300. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, M.; Wang, A.J.; Chen, Y.; Zhao, G.; Jiang, Z.; Wang, X.; Shi, D.; Zhang, T.; Sun, B.; He, H.; et al. Efficacy and safety of erythropoietin for traumatic brain injury. BMC Neurol. 2020, 20, 399. [Google Scholar] [CrossRef] [PubMed]
  124. Jha, R.M.; Kochanek, P.M.; Simard, J.M. Pathophysiology and treatment of cerebral edema in traumatic brain injury. Neuropharmacology 2019, 145, 230–246. [Google Scholar] [CrossRef] [PubMed]
  125. Ballabh, P.; Braun, A.; Nedergaard, M. The blood–brain barrier: An overview: Structure, regulation, and clinical implications. Neurobiol. Dis. 2004, 16, 1–13. [Google Scholar] [CrossRef]
  126. Mira, R.G.; Lira, M.; Cerpa, W. Traumatic Brain Injury: Mechanisms of Glial Response. Front. Physiol. 2021, 12, 740939. [Google Scholar] [CrossRef] [PubMed]
  127. Donkin, J.J.; Vink, R. Mechanisms of cerebral edema in traumatic brain injury: Therapeutic developments. Curr. Opin. Neurol. 2010, 23, 293–299. [Google Scholar] [CrossRef]
  128. Minta, K.; Brinkmalm, G.; Al Nimer, F.; Thelin, E.P.; Piehl, F.; Tullberg, M.; Jeppsson, A.; Portelius, E.; Zetterberg, H.; Blennow, K.; et al. Dynamics of cerebrospinal fluid levels of matrix metalloproteinases in human traumatic brain injury. Sci. Rep. 2020, 10, 18075. [Google Scholar] [CrossRef]
  129. Vink, R.; Gabrielian, L.; Thornton, E. The Role of Substance P in Secondary Pathophysiology after Traumatic Brain Injury. Front. Neurol. 2017, 8, 304. [Google Scholar] [CrossRef]
  130. Gao, X.; Bayraktutan, U. Substance P reversibly compromises the integrity and function of blood-brain barrier. Peptides 2023, 167, 171048. [Google Scholar] [CrossRef] [PubMed]
  131. Hadanny, A.; Maroon, J.; Efrati, S. The Efficacy of Hyperbaric Oxygen Therapy in Traumatic Brain Injury Patients: Literature Review and Clinical Guidelines. Med. Res. Arch. 2023, 11. [Google Scholar] [CrossRef]
  132. Hu, Q.; Manaenko, A.; Xu, T.; Guo, Z.; Tang, J.; Zhang, J. Hyperbaric Oxygen Therapy for Traumatic Brain Injury: Bench-to-Bedside. Med. Gas. Res. 2016, 6, 102. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The summarized pathophysiology of TBI. This review will be separated into two parts. Part I will focus on the cellular and Brain blood barrier (BBB) that evolve following the injury. Part II will focus on the consequence of these molecular mechanisms and its repair.
Figure 1. The summarized pathophysiology of TBI. This review will be separated into two parts. Part I will focus on the cellular and Brain blood barrier (BBB) that evolve following the injury. Part II will focus on the consequence of these molecular mechanisms and its repair.
Biomedicines 12 00520 g001
Figure 2. Schematic view of the primary, secondary, and tertiary injuries after TBI. Solid arrows indicate an association, and dashed arrows indicate the following stage (leads to).
Figure 2. Schematic view of the primary, secondary, and tertiary injuries after TBI. Solid arrows indicate an association, and dashed arrows indicate the following stage (leads to).
Biomedicines 12 00520 g002
Figure 3. Schematic evolution of the major mechanisms involved in the secondary injury of TBI. The sequence of events can occur simultaneously and interact to exacerbate injury and initiate neuronal and vascular repair. Potential drugs are placed in red-doted arrows directed to their leading site of action. Black solid lines indicate association, while red continuous indicate an inhibitor of the pathway.
Figure 3. Schematic evolution of the major mechanisms involved in the secondary injury of TBI. The sequence of events can occur simultaneously and interact to exacerbate injury and initiate neuronal and vascular repair. Potential drugs are placed in red-doted arrows directed to their leading site of action. Black solid lines indicate association, while red continuous indicate an inhibitor of the pathway.
Biomedicines 12 00520 g003
Table 1. Main cytokines 1 involved in traumatic brain injury.
Table 1. Main cytokines 1 involved in traumatic brain injury.
Main Effects in TBIHighlights
IL1βDetrimentalRaised CSF IL1β correlates with both raised ICP and poorer outcome. The balance between members of the IL1 cytokine family, in particular between IL1β and its endogenous inhibitor IL1ra, is an important determinant of the degree of inflammatory response, rather than the absolute concentration of IL1β. In TBI patients, high microdialysate IL1ra/IL1b ratio is associated to favourable outcomes [54,55,56].
TNFαDetrimental in acute phase and beneficial during the healing processUpregulated in the injured brain early after trauma, reaching a peak within a few hours following the initial injury. This cytokine triggers the apoptotic cascade but also, pathways resulting in activation of pro-survival genes [57,58,59].
IL6BeneficialA trophic factor that is upregulated in the CNS after injury and promotes neuronal survival and brain repair through astroglia and vascular remodelling. Following TBI, its concentration rises dramatically [60,61].
ComplementDetrimentalThe complement system significantly triggers inflammation in TBI, increasing BBB permeability and inducing cytokines, chemokines, and adhesion molecules. Key products like C3a and C5a enhance vascular permeability and inflammation. The cascade forms the membrane attack complex (MAC), affecting both non-nucleated and nucleated cells. C5a, a major pro-inflammatory component, amplifies cytokine production and leukocyte adhesion, leading to more inflammation. This system also contributes to apoptosis and cell death in TBI [62,63,64].
AngiopoietinsBeneficial (Ang1) and detrimental (Ang2)They are family of growth factors that are important in regulating angiogenesis and vascular permeability, and also have been implicated in BBB disruption. TBIs models show acute decrease in Angiopoetin-1 expression and concomitant increase in Angiopoetin-2 which is associated with endothelial apoptosis and BBB permeability [65,66,67,68,69].
1 IL1β: interleukin1β; TNFα: tumor necrosis factor α; IL6: interleukin6.
Table 2. Summary of Potential Therapeutic Targets and Drugs for TBI.
Table 2. Summary of Potential Therapeutic Targets and Drugs for TBI.
PathophysiologyTarget/DrugClinical/Preclinical EvidenceKey Findings/Comments
Excitotoxicity and CalciumStatinsCultured neurons protection from NMDA-induced death [31]Potential protection, contradictory clinical results
AmantadineAccelerates functional recovery in TBI patients [32]Beneficial for prolonged disorders of consciousness
Nimodipine (Calcium channel blocker)Contradictory results in clinical trials [36]Some argue hindrance of synaptic transmission
Magnesium SulfatePromising in experimental studies, clinical efficacy TBD [37,38]Potential effectiveness in TBI treatment
Free Radicals and Oxidative StressCiticolineMultiple neuroprotective mechanisms [45], no benefit in RCT [46]Inhibition of oxidative stress and apoptotic pathways
StatinsUpregulate eNOS, reduce hypoperfusion, protect neurons [46]Reduction in mortality risk in TBI individuals
Inflammatory Mediators and CascadesInnovative Anti-inflammatory DrugsTargeting NF-κB, MAPKs, JAK/STAT, PI3K/Akt/mTOR, TGF-β1 [74]Potential for reducing apoptosis and inflammation
Recombinant ErythropoietinIncreases JAK and STAT, reduces apoptosis in TBI [79]Potential for neuroprotection
MAPK Pathway (p38, JNK)Activation contributes to increased neuronal damage [80]Inhibition may reduce neuronal damage
TNF and iNOSDual nature of inflammation, complex effects [82]TNF associated with brain edema, iNOS inhibition worsens spatial memory [85,86,87]
Other Neuroinflammatory ComponentsIL-1 Receptor InhibitorsInhibit inflammasome activation, reduce IL-1β release [96]Potential to limit neuroinflammation
Intravenous ImmunoglobulinInhibit priming of T cells from entering CNS [97]Potential inhibition of T cell infiltration
NSAIDsInhibition of COX, reduction in IL-1β, anti-inflammatory [100]Modulation of proinflammatory pathways
Mitochondrial DysfunctionCyclosporine A (CsA)Stabilizes mPTP, neuroprotection in preclinical TBI [102]Reduction in lesion volume in animal models
Therapeutic HypothermiaMixed results in meta-analysis, potential benefits in specific cases [103]Not universally beneficial, effective in increased intracranial pressure
BarbituratesSuppress cerebral metabolism, reduce ICP [114]Significant ICP decrease but accompanied by instability
Cell DeathErythropoietin (Epo)Modulates caspase activities, mitochondrial function [124]Potential for neuroprotection, modulation of cell death
BBB Disruption and Neutrophil InvasionSubstance P AntagonistsLimit BBB opening, edema after TBI [130]Promising results, potential BBB protection
Hyperbaric Oxygen Therapy (HBOT)Reduces MMP-9, inhibits neutrophilic infiltration [132]Potential benefits in acute moderate-severe TBI
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

de Macedo Filho, L.; Figueredo, L.F.; Villegas-Gomez, G.A.; Arthur, M.; Pedraza-Ciro, M.C.; Martins, H.; Kanawati Neto, J.; Hawryluk, G.J.; Amorim, R.L.O. Pathophysiology-Based Management of Secondary Injuries and Insults in TBI. Biomedicines 2024, 12, 520. https://doi.org/10.3390/biomedicines12030520

AMA Style

de Macedo Filho L, Figueredo LF, Villegas-Gomez GA, Arthur M, Pedraza-Ciro MC, Martins H, Kanawati Neto J, Hawryluk GJ, Amorim RLO. Pathophysiology-Based Management of Secondary Injuries and Insults in TBI. Biomedicines. 2024; 12(3):520. https://doi.org/10.3390/biomedicines12030520

Chicago/Turabian Style

de Macedo Filho, Leonardo, Luisa F. Figueredo, Gustavo Adolfo Villegas-Gomez, Matthew Arthur, Maria Camila Pedraza-Ciro, Henrique Martins, Joaquim Kanawati Neto, Gregory J. Hawryluk, and Robson Luís Oliveira Amorim. 2024. "Pathophysiology-Based Management of Secondary Injuries and Insults in TBI" Biomedicines 12, no. 3: 520. https://doi.org/10.3390/biomedicines12030520

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