Next Article in Journal
The Influence of a Personalized Intervention Program—AGA@4life—in the Cardiovascular Diseases: A Biochemical Approach
Previous Article in Journal
A Comprehensive Literature Review on Diagnostic Strategies and Clinical Outcome of Intraoral Angiosarcoma and Kaposi Sarcoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury

by
Zahra Hasanpour-Segherlou
1,*,
Forough Masheghati
2,
Mahdieh Shakeri-Darzehkanani
2,
Mohammad-Reza Hosseini-Siyanaki
3 and
Brandon Lucke-Wold
1
1
Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
2
Medicine Faculty, University of Tabriz, Tabriz 51656-87386, Iran
3
Department of Radiology, University of Florida, Gainesville, FL 32608, USA
*
Author to whom correspondence should be addressed.
J. Vasc. Dis. 2024, 3(3), 319-332; https://doi.org/10.3390/jvd3030025
Submission received: 27 May 2024 / Revised: 16 August 2024 / Accepted: 4 September 2024 / Published: 6 September 2024
(This article belongs to the Section Neurovascular Diseases)

Highlights

What are the main findings?
  • Traumatic Brain Injury (TBI) causes vascular damage, leading to blood–brain barrier (BBB) disruption, neuroinflammation, and ischemia, all contributing to long-term neurodegeneration;
  • TBI-related cerebrovascular changes, such as BBB breakdown, coagulopathy, and microhemorrhages, are linked to dementia and other neurodegenerative diseases;
  • Sex differences influence TBI outcomes, with hormonal and cellular responses varying, potentially affecting recovery.
What is the implication of the main finding?
  • Understanding the vascular implications of TBI provides insight into managing long-term neurodegenerative risks;
  • Targeted interventions for BBB integrity and immune modulation could mitigate the chronic effects of TBI and improve patient outcomes;
  • Recognizing sex-based differences may inform personalized approaches to TBI treatment and neuroprotection.

Abstract

:
Traumatic brain injury (TBI) results from external biomechanical forces that cause structural and physiological disturbances in the brain, leading to neuronal, axonal, and vascular damage. TBIs are predominantly mild (65%), with moderate (10%) and severe (25%) cases also prevalent. TBI significantly impacts health, increasing the risk of neurodegenerative diseases such as dementia, post injury. The initial phase of TBI involves acute disruption of the blood–brain barrier (BBB) due to vascular shear stress, leading to ischemic damage and amyloid-beta accumulation. Among the acute cerebrovascular changes after trauma are early progressive hemorrhage, micro bleeding, coagulopathy, neurovascular unit (NVU) uncoupling, changes in the BBB, changes in cerebral blood flow (CBF), and cerebral edema. The secondary phase is characterized by metabolic dysregulation and inflammation, mediated by oxidative stress and reactive oxygen species (ROS), which contribute to further neurodegeneration. The cerebrovascular changes and neuroinflammation include excitotoxicity from elevated extracellular glutamate levels, coagulopathy, NVU, immune responses, and chronic vascular changes after TBI result in neurodegeneration. Severe TBI often leads to dysfunction in organs outside the brain, which can significantly impact patient care and outcomes. The vascular component of systemic inflammation after TBI includes immune dysregulation, hemodynamic dysfunction, coagulopathy, respiratory failure, and acute kidney injury. There are differences in how men and women acquire traumatic brain injuries, how their brains respond to these injuries at the cellular and molecular levels, and in their brain repair and recovery processes. Also, the patterns of cerebrovascular dysfunction and stroke vulnerability after TBI are different in males and females based on animal studies.

1. Introduction

Traumatic brain injury (TBI) occurs when external biomechanical forces cause disturbances in the structure and physiology of the brain due to damage to the skull, leading to neuronal, axonal, and vascular injuries [1]. The majority of TBIs are categorized as mild (65%), with 10% classified as moderate and 25% as severe [2]. TBI has a huge health significance in societies and may increase the risk of neurodegenerative pathology by different mechanisms [3,4,5]. It can trigger dementia and lead to neurodegeneration [6,7]. Studies suggest a robust connection between developing neurodegenerative disorders and moderate to severe TBI, revealing a two to four times elevated risk of dementia. Moreover, experiencing a mild traumatic brain injury (MTBI) or another form of TBI can elevate the risk of developing dementia, persisting up to 30 years from the initial injury [8]. One cross-sectional analysis indicated that a history of TBI, even in the absence of loss of consciousness (LOC), is correlated with age-related neuropathological outcomes, having both neurodegenerative and vascular aspects [9]. Specifically, TBI with a LOC lasting more than 1 h was found to be associated with an elevated risk of cortical microinfarcts in studies such as the Religious Orders Study and Memory and Aging Project [10]. A recent meta-analysis revealed a 70% increased risk of dementia associated with TBI, especially in Asians and young men [11]. Another systematic review showed that there was evidence of dementia in TBI patients, reported in 15.5% of 44 published papers [12]. The triggering mechanisms for secondary neurodegeneration following TBI involve a sequence of interconnected pathological changes induced by the initial traumatic injury [1]. TBI can trigger acute disruption of the blood–brain barrier (BBB) through vascular shear stress, leading to both ischemic damage and the accumulation of Aβ [13,14]. The processes of hypoperfusion, vascular dysfunction, and ischemia following TBI may collectively contribute to Aβ deposition, playing a role in the secondary injury including endothelial cell dysfunction, cerebrovascular damage, oxidative stress, and mitochondrial damage [15].
Traumatic cerebrovascular injury is one of the most common abnormalities acutely reported in TBI patients [16]. Cerebral vascular dysfunction may occur before neuronal damage or develop as a consequence of neuronal dysfunction [17]. The cerebrovascular system’s pathology after TBI may originate from either direct injury to its components or as a result of a subsequent pathophysiological cascade triggered after the initial injury [17]. The dysfunction in blood vessel function seen immediately after the initial TBI persists for many months and years [18]. Further investigation is needed to explore the correlation between cerebrovascular pathology, neuroinflammation, and the severity of trauma [15].
The acute phase of TBI, typically described as the first week following injury, is characterized by predominant mechanical damage [19]. Initially, TBI disrupts the BBB, allowing activated leukocytes to migrate into the damaged brain tissue [19]. This mechanism is aided by the increased expression of cell adhesion molecules [19]. Macrophages become apparent immediately after the migration of polymorphonuclear leukocytes (PMNs), even though resident intracerebral macrophages (microglia) are uniformly distributed throughout the central nervous system [20]. Activated astrocytes, microglia, and leukocytes are recognized as sources of the pro-inflammatory cytokine response. They generate inflammatory molecules (e.g., cytokines and chemokines) and reactive oxygen species (ROS), which play a role in demyelination and the disturbance of the axonal cytoskeleton [19]. This disruption leads to axonal swelling and the buildup of transport proteins at the terminals, thus impairing the activity of neurons [19]. The ongoing axonal damage ultimately causes neurodegeneration [19]. Some of acute cerebrovascular changes after trauma are early progressive hemorrhage, micro bleeding, coagulopathy, neurovascular unit (NVU) uncoupling, changes in the BBB, changes in cerebral blood flow (CBF), and cerebral edema.
The secondary phase of injury, which can last months to years, is recognized by the delayed onset of metabolic dysregulation and inflammatory pathways [21,22,23]. Oxidative stress, which is characterized by ROS, seems to mediate the secondary phase of injury [24,25,26]. ROS are produced as a result of glutamate excitotoxicity, mitochondrial dysfunction, and endoplasmic reticulum stress [24,25,26]. ROS trigger lipid peroxidation and cause the disruption of the cell membrane; they also cause protein damage, which then leads to neurodegeneration and neuroinflammation [27]. The oxidative damage leads to an increase in pro-inflammatory cytokines (e.g., TNF-α, IL-1α, IL-1β, and IL-6) and causes further neurodegeneration [28]. TBI results in the cell accumulating Ca2+, which activates many widely distributed enzymes such as nitric oxide synthase, xanthine dehydrogenase, and phospholipase A2 that increase NO and O2 production [29]. The cascade can lead to structural changes in the membrane and in mitochondria, lipids, proteins, and DNA [30].

2. Excitotoxicity

Some studies have identified a number of cellular and molecular mechanisms that make a contribution to the development of cytotoxic and vasogenic edema, including the degradation of BBB components by matrix metalloproteinases, excitotoxicity due to excessive glutamate release, ion pump failure, mitochondrial dysfunction, the inflammation-induced release of vasoactive agents, the insertion of aquaporin 4 water channels into the cell membrane increasing the bi-directional flow of water driven by the net Starling force, and mechanical injury to the vasculature and tissue [31,32].
The extracellular accumulation of glutamate leads to an overstimulation of glutamate receptors, causing damage to both neurons and glial cells through excitotoxicity [33]. Increased levels of extracellular glutamate following TBI have been demonstrated to play a role in causing additional harm in experimental studies [34,35,36,37,38]. Recent research has shown a notable decrease in glial glutamate transporter proteins following experimental TBI, leading to elevated levels of extracellular glutamate [39,40,41]. Astrocytes play a key role in removing glutamate released at synapses through the primary mechanism of glutamate uptake [42,43]. A study showed that within the initial 24 h after TBI, the appearance of cells expressing the glutamate transporter at the site of contusion underwent morphological changes [44]. Numerous cells were massively swollen [44]. This morphology was also the most common during the 24 h to 7-day survival period [44]. In the later stages of survival, cells that were positive for excitatory amino acid transporters showed an increase in quantity and displayed characteristics such as highly stained, moderately branched processes and hypertrophic cytoplasm, resembling the appearance of reactive astrocytes [44]. After 7 days post injury, various astrocyte shapes were observed together at the contusion site [44]. The number of these cells was reduced significantly after three months post injury [44]. Astrocyte activation resulting from TBI is referred to as astrogliosis [45]. This phenomenon involves an atypical elevation in the quantity of astrocytes, changes in gene expression, alterations in morphology, and the formation of scar tissue [45]. Astrocytes exhibit signs of hypertrophy within 2 to 3 days following injury, and this hypertrophic state persists for up to 7 days post injury, concomitant with the formation of a glial scar [46]. Reactive astrogliosis has the capacity to persist for a duration of 60 days [46].

3. Coagulopathy and Hemorrhage

Patients who sustained a traumatic intracranial hemorrhage remained at risk for developing coagulopathy until 72 h after the trauma [47]. TBI coagulopathy is frequently broadly defined as any disturbance in a patient’s coagulation parameters [48]. Coagulopathy can emerge up to 5 days after injury, with its incidence showing a linear association with the severity of the injury [49]. The exact mechanisms responsible for coagulopathy in TBI have not yet been fully identified [50]. The coagulation pathway may result in disseminated intravascular coagulation (DIC) [50]. Patients with coagulopathy after TBI have poorer outcomes than others [51].
Early progressive hemorrhage occurs in approximately 50% of head-injured patients who undergo CT scanning within 2 h of injury; it occurs most frequently in cerebral contusions, and it is correlated with ICP elevations [52]. Microbleeds have been found in both the acute and chronic phases after TBI [53,54,55]. These accumulations of ferritin/hemosiderin are recognized to be harmful to endothelial cells and astrocytes [56,57], which can start cascades of inflammation. The cascades may involve the stimulation of gliosis and activation of apoptosis that have been linked to post-traumatic neurodegenerative outcomes [58]. Inflammation may persist for years following a single TBI [59]. Furthermore, microvascular abnormalities linked to axonal changes have been observed for up to 3 weeks following experimental TBI [60,61,62,63].

4. NVU Uncoupling

NVU is a vessel-centered concept, emphasizing that all cellular components play an integrated role in maintaining the normal physiological functions of the brain [64]. The neurovascular unit is formed by the neuron and its supporting cells, including astrocytes, endothelial cells, pericytes, and smooth muscle cells [65]. Their primary role is to provide various nutrients, including those required for nerve structure and distribution [64]. After brain injury, glial cells primarily regulate nerve repair factors and significantly contribute to the regeneration of both the central and peripheral nervous systems [66]. Their primary function involves integrating and interpreting signals released from adjacent cells, which leads to various functional outcomes such as modulating BBB permeability, promoting angiogenesis, eliminating toxic metabolites, regulating capillary blood flow dynamics, attenuating nerve inflammation, and modulating stem cell activity [67]. Hypoxia of the tissue frequently serves as the primary stimulus for a series of pathophysiological alterations within the NVU [68]. The changes following hypoxia include the disruption of interendothelial tight junctions, retraction of pericytes from the abluminal surface of the capillary, breakdown of the basal lamina with transudation of plasma, infiltration of inflammatory cells, endothelial cell proliferation and migration, and in some cases, vasculogenesis [68]. At the molecular level, this reorganization is accompanied by the increased expression of endothelial cell leukocyte adhesion receptors, loss of endothelial cell and astrocyte integrin receptors, loss of their matrix ligands, expression of members of several matrix-degrading protease families, and the appearance of receptors associated with angiogenesis [69]. NVU injury triggers the loss of BBB function and neurovascular uncoupling [68], marking the first pathological steps involved in initiating neurodegenerative cascades, and gradually lead to neurodegeneration [70].

5. BBB Disruption

The BBB is composed of the endothelial tight junction complex, which seals the intracellular space between endothelial cells and regulates transcellular transport [17]. One of the most serious outcomes of TBI is the disruption of the BBB, which varies according to patients’ age and the type and severity of the injury [71,72]. The immediate pathological change in the BBB following TBI is the disruption of its tight junctions, leading to an increase in paracellular permeability [73]. The extravasation of serum proteins such as fibrinogen and immunoglobulin G, both markers for BBB disruption, is observed in the brains of human patients who died in the acute phase following TBI, as well as in those who survived for at least 12 months [73].
The loss of pericytes is a key indicator of dysfunction in the BBB and has been proposed to initiate various pathological issues like abnormal BBB permeability, micro-aneurysm development, ischemia, and other related conditions [74,75]. It was shown that the initiation of oxidative stress triggers the activation of matrix metalloproteinases (MMPs), resulting in the breakdown of the BBB and the initiation of inflammatory responses after TBI [76,77]. Another research study showed that MMP-9 causes pericytes to move away from the endothelium, resulting in pericyte loss and disruption of the BBB [78]. PDGFR-β, αSMA (alpha-smooth muscle actin), NG2, CD13, and desmin are commonly used markers for pericytes [42,75,79,80]. PDGFR-β seems to be the most specific and is highly expressed in pericytes [81]. A study indicated that the levels of PDGFR-β, NG2, and CD13 were notably decreased after TBI [82]. However, the level of PDGFR-β expression decreased significantly at 12 h after TBI, then gradually increased at 24 h and 48 h, but did not reach the same level as in the uninjured samples [82]. The recruitment of CNS pericytes is dependent on the signaling of PDGF-B/PDGFR-β [83,84]. The PDGF-B secreted by endothelial cells binds to the PDGFR-β on pericytes, triggering various signaling pathways that control the growth, migration, and attraction of pericytes to the blood vessel wall [85,86]. Bhowmick et al.’s study indicated that the decrease in PDGFR-β expression on pericytes due to injury impacted the expression of PDGF-B on endothelial cells, leading to a disruption in PDGF-B/PDGFR-β signaling [82]. Also, they observed a decrease in the levels of N-cadherin and connexin-43 after TBI [82]. As pericytes and endothelial cells are linked through N-cadherin and connexin-43 in the neurovascular unit, the integrity of the BBB is clearly impacted by the absence of pericytes [82].

6. CBF Changes

Neuroimaging studies consistently demonstrate reduced CBF shortly after experimental TBI [17]. One study showed the changes in CBF in both cortical and subcortical regions using ASL-MRI, which may underlie deficits in cognitive function observed in MTBI patients during the acute stage [87]. In one study, it was demonstrated that patients whose CBF returns to normal levels at 2–3 weeks following traumatic brain injury, after being abnormally low in the acute phase of injury, can be expected to achieve a good neurological outcome [88].
Cerebral edema and the subsequent elevated ICP are linked to mortality and poor prognosis after TBI [89]. Increased ICP is linked to coma, brain herniation, and death [90]. Marmarou et al. [91] diagnosed that edema drives brain swelling in human TBI, as opposed to vascular engorgement and an increase in cerebral blood volume. Cerebrovascular injuries following TBI, including microbleeds and CBF dysfunction, contribute to poor outcomes and neurodegeneration [92].

7. Immune Reaction

Key contributors to the inflammatory response following brain injury include immune cells from the peripheral blood, such as neutrophils, as well as resident immune cells like macrophages and microglia [93,94,95,96,97]. All of these cells have the capability to generate pro-inflammatory substances, such as chemokines, the significant involvement of which in brain damage has been definitively demonstrated [98,99,100]. Resident microglia in the affected area and peripheral macrophages are promptly activated and migrate to the site of injury, where they begin to release signaling molecules and attract additional immune cells [101,102]. Distinguishing between the functions of resident microglia and infiltrating peripheral macrophages is a current focus of research [103]. This similarity complicates the differentiation of their respective roles in intracerebral inflammation [103]. The signature genes M1 (classically activated macrophage) and M2 (alternatively activated macrophage) are not exclusively expressed in microglia/macrophages, but also in other cells within the CNS or immune cells that have infiltrated the area [104]. A study showed that the presence of the M1 marker CD16/32 showed a slight increase in the cortex one day after TBI, and then significantly increased until day 14, and surprisingly, there were more CD16/32-positive microglia/macrophages in the opposite cortex three days after TBI compared to mice that underwent a sham operation [104]. Immunofluorescence for the M2 marker CD206 showed a notable increase above normal levels in the cortex and striatum on the same side as the injury three days after TBI [104]. The levels peaked on day 5 and then returned to baseline by day 14 [104]. These results show that there is a change over time in the type of microglial cells present after a mechanical injury, transitioning from a temporary M2 phenotype to a lasting M1 phenotype within a 14-day period [104]. The long-term impairment in myeloid and lymphocyte reactions following TBI indicates that TBI can lead to lasting changes in peripheral immune cell function [105]. The study revealed that there were notable deficiencies in the polarization of bone marrow-derived macrophages (BMDMs) obtained from mice with chronic TBI in comparison to BMDMs from sham control mice of equivalent time points [105]. Hence, BMDMs in mice with chronic TBI demonstrate permanent changes in the expression of inflammatory genes, potentially influenced by underlying epigenetic processes [105].
TBI triggers the activation of resident microglia, leading to the production of cytokines. This results in the entry of immune cells from the peripheral system, followed by a sustained activation of the brain’s resident microglia and astrocytes [93,106,107]. They are one of the initial groups to react to brain injury, and their continuous activation in animal models and individuals with TBI could play a role in causing long-term functional impairments [97,108,109,110,111]. Research conducted on spinal cord and ischemic brain injury models has revealed that most microglia and macrophages present at the injury site exhibit a combination of M1 and M2 activation characteristics [112,113]. However, the M2 response is temporary, and within a week of the injury, there is a transition towards a predominant M1-like response [112,113]. Furthermore, microglial cell phenotypes can vary based on tissue type, with a quicker shift from M2- to M1-dominant phenotypes observed in the white matter compared to the gray matter following TBI [104].

8. Chronic Vascular Changes after TBI

Angiogenesis is a key component of adult vascular remodeling and is facilitated by both mature endothelial cells and endothelial progenitor cells (EPCs) [114]. After TBI, EPCs can be found in the bone marrow and peripheral blood, from where they are moved to the peripheral blood [114]. After TBI, the number of CD34+ EPCs in peripheral blood increases at 24 h, peaks at 48 h, and returns to a normal level by 168 h [114]. As soon as 24 h after TBI, CD34+ cells can be identified in the area surrounding the damaged brain, and by 72 h after TBI, a CD34+ endothelial-like cell lining can be seen in the vessel-lumen structure [114]. The bone marrow-derived EPCs support endothelial repair and contribute to the formation of new blood vessels in ischemic conditions [115]. Vascular endothelial growth factor (VEGF) is recognized for its significant role in the process of angiogenesis [116]. Elevated levels of VEGFR1 and VEGFR2 were also observed in a cortical impact model of TBI [117].

9. Systemic Inflammation after TBI

Severe TBI often leads to dysfunction in organs outside the brain, which can significantly impact patient care and outcomes [118,119]. The effects of this extracranial multiorgan dysfunction on the brain include decreased cerebral blood flow, cerebral hypoxia, acidosis, altered metabolism, and bleeding. These factors can contribute to secondary brain injury and worsen clinical outcomes [118,119]. In addition to primary brain damage, secondary brain injuries worsen the outcomes of TBI [120,121]. The most common extracranial complications include multiple organ failure and infections [120,121]. The vascular component of systemic inflammation after TBI includes immune dysregulation, hemodynamic dysfunction, coagulopathy, respiratory failure, and acute kidney injury [120]. Cardiac dysfunction is a reported consequence of TBI, particularly common in patients with more severe injuries, which causes hypotension, CBF changes, and secondary brain injuries [122,123]. Its hemodynamic and histologic profile suggests that sympathetic activation may be the underlying cause [120,124,125]. Recent studies have refined blood pressure thresholds for optimizing outcomes in severe TBI, considering age-dependent variations [120]. Significantly, cardiac dysfunction often develops shortly after the injury, a time when the injured brain is especially vulnerable to reduced cerebral blood flow [122]. However, cardiac function typically shows improvement during the first week of hospitalization after a traumatic brain injury [122]. Inappropriate activation of coagulation pathways may worsen organ dysfunction following severe TBI and is closely linked to negative outcomes, such as higher mortality rates, extended periods of mechanical ventilation, and increased functional disability [126,127,128]. Meta-analyses show that about one-third of TBI patients, particularly those with severe TBI, develop coagulopathy [129,130]. Ultimately, maladaptive coagulopathy manifests as DIC syndrome, resulting in microthrombosis, bleeding, and secondary brain injuries from both hemorrhagic and ischemic causes [131].

10. Sex Difference in TBI

There are differences in how men and women acquire traumatic brain injuries, how their brains respond to these injuries at the cellular and molecular levels, and in their brain repair and recovery processes [132]. In TBI, extracerebral trauma may follow sex-specific patterns and lead to varying post-injury complications, which can result in different outcomes between sexes [132]. Women are more likely to experience injuries from assault or violence within interpersonal relationships, while men are more prone to work-related injuries due to falls and motor vehicle collisions [133,134,135]. Sex hormones and genetics probably impact edema, inflammation, excitotoxicity, oxidative stress, and mitochondrial function in both males and females following TBI [132]. Research also suggests that there are sex-based differences in recovery processes and brain repair, with varying levels of neuroplasticity observed between genders [132]. Given the current complex research on TBI in both males and females, it is not possible to definitively determine that one sex has better outcomes than the other [132,136]. The effect of sex hormones on vascular pathology after TBI has been studied in animal models. A study using a mouse model has demonstrated that following a mild TBI, male and female mice experience different patterns of cerebrovascular dysfunction and stroke vulnerability over time [137]. Sex differences in stroke vulnerability may partly result from previous brain injuries, which cause lasting changes in intravascular coagulation, blood–brain barrier permeability, and angiogenesis [137]. Another study using juvenile male mice demonstrated less acute inflammatory cytokine expression, greater subacute microglial/macrophage accumulation, and better neurological recovery compared to females, highlighting the importance of considering age and sex in TBI research to identify novel therapeutic targets [138]. In a rat model of TBI, male rats showed increased edema peaking at 5 h post injury, while female rats peaked at 24 h, with ovariectomized females displaying a profile similar to males. These differences suggest that endogenous hormone levels influence edema formation after TBI, potentially impacting clinical management [139].
Progesterone, a developmental hormone with neurosteroidal properties, has demonstrated neuroprotective and functional efficacy in numerous laboratories globally, across various animal models of TBI and stroke, leading to improved outcomes [140,141,142,143]. A study revealed that female rats in the proestrus stage of the estrous cycle (equivalent to the follicular phase in women) exhibit significantly smaller cortical contusion volumes compared to ovariectomized rats [144]. These proestrus rats also demonstrate a greater reduction in depression and show better behavioral outcomes following TBI [145]. In addition to research demonstrating the influence of estrous phases on TBI outcomes, studies have confirmed that TBI also disrupts estrous cyclicity in animal models, mirroring the post-injury menstrual disruption observed in women [146,147]. In experimental TBI models, post-injury estrous cyclicity is impaired, with animals spending more time in the diestrus phase (diestrus and metestrus are phases when progesterone levels peak) [146,147].
A meta-analysis of eight studies examining gender differences in TBI outcomes across all severity levels found that women had worse outcomes in 17 out of the 20 variables assessed [148]. Other studies have found that women, particularly those who are premenarchal or postmenopausal, tend to have better post-TBI outcomes compared to men [149,150]. These variables included postconcussive symptom scores, length of hospitalization, cognitive decline, depression, headache, return to work, reaction time compared to baseline, and death [151]. The phase of the menstrual cycle appears to be linked to quality of life and neurological outcomes after MTBI. Women who sustain an injury during the luteal phase, when progesterone levels are highest, experience worse outcomes compared to those in the follicular phase, when progesterone is low, or women taking synthetic progestins [152]. These findings seem to support the hypothesis that the acute withdrawal of progesterone following MTBI may play a role in the gender differences observed after the injury [151]. Conversely, a study found that women often miss menstrual periods or experience complete amenorrhea after TBI, indicating potential post-injury deficits in sex hormones [153]. Other studies have more conclusively demonstrated that TBI leads to the suppression of the hypothalamic–pituitary–gonadal axis, resulting in reduced levels of luteinizing hormone, follicle-stimulating hormone, testosterone, estrogen, and progesterone [154]. Although the preclinical documentation and multiple positive Phase II clinical trials were generally favorable, Phase III clinical trials revealed that progesterone was ineffective for patients with moderate to severe TBI [140].

11. Conclusions

TBI initiates a cascade of acute and chronic pathological processes, beginning with immediate mechanical damage and BBB disruption, followed by inflammation and neurodegeneration. The acute phase involves leukocyte migration into damaged brain tissue, releasing pro-inflammatory cytokines and ROS. The secondary phase extends over months to years, marked by oxidative stress, metabolic dysregulation, and continued inflammation. Elevated extracellular glutamate levels cause excitotoxicity, damaging neurons and glial cells. Coagulopathy and hemorrhage complicate recovery, with risks persisting up to 72 h post injury. NVU injury disrupts BBB integrity, leading to neurodegenerative cascades. Immune responses, including microglial activation and peripheral immune cell infiltration, play a significant role in sustaining inflammation and functional impairments. Chronic vascular changes, such as angiogenesis and endothelial repair, are crucial in the progression of TBI-induced neurodegeneration. Severe TBI often leads to dysfunction in organs outside the brain, which can significantly impact patient care and outcomes. The vascular component of systemic inflammation after TBI includes immune dysregulation, hemodynamic dysfunction, coagulopathy, respiratory failure, and acute kidney injury. There are differences in how men and women acquire traumatic brain injuries, how their brains respond to these injuries at the cellular and molecular levels, and in their brain repair and recovery processes. Also, the patterns of cerebrovascular dysfunction and stroke vulnerability after TBI are different in males and females based on animal studies. A comprehensive understanding of these mechanisms is essential for developing targeted interventions to mitigate long-term effects and improve outcomes for TBI patients. As a future direction, controlling the vascular component of systemic inflammation and understanding organ cross-talk post-TBI could address the broader impacts on patient health. Also, it might be helpful to examine the role of estrogen in protecting against vascular changes after TBI.

Author Contributions

Conceptualization, B.L.-W.; writing, F.M., M.S.-D. and M.-R.H.-S.; review and editing, Z.H.-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.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dodd, W.S.; Panther, E.J.; Pierre, K.; Hernandez, J.S.; Patel, D.; Lucke-Wold, B. Traumatic Brain Injury and Secondary Neurodegenerative Disease. Trauma Care 2022, 2, 510–522. [Google Scholar] [CrossRef]
  2. Brennan, D.; Delaney, C.; Farrell, M.; Campbell, M.; Doherty, C.P. Polypathology-associated neurodegeneration after remote head injury. Clin. Neuropathol. 2023, 42, 201–211. [Google Scholar] [CrossRef] [PubMed]
  3. Critchley, M. Medical aspects of boxing, particularly from a neurological standpoint. Br. Med. J. 1957, 1, 357. [Google Scholar] [CrossRef]
  4. Martland, H.S. Punch drunk. J. Am. Med. Assoc. 1928, 91, 1103–1107. [Google Scholar] [CrossRef]
  5. Millspaugh, J. Dementia pugilistica. US Nav. Med. Bull. 1937, 35, e303. [Google Scholar]
  6. Sariaslan, A.; Sharp, D.J.; D’Onofrio, B.M.; Larsson, H.; Fazel, S. Long-term outcomes associated with traumatic brain injury in childhood and adolescence: A nationwide Swedish cohort study of a wide range of medical and social outcomes. PLoS Med. 2016, 13, e1002103. [Google Scholar] [CrossRef] [PubMed]
  7. Scheid, R.; Walther, K.; Guthke, T.; Preul, C.; von Cramon, D.Y. Cognitive sequelae of diffuse axonal injury. Arch. Neurol. 2006, 63, 418–424. [Google Scholar] [CrossRef] [PubMed]
  8. El-Menyar, A.; Al-Thani, H.; Mansour, M.F. Dementia and traumatic brain injuries: Underestimated bidirectional disorder. Front Neurol 2023, 14, 1340709. [Google Scholar] [CrossRef]
  9. Agrawal, S.; Leurgans, S.E.; James, B.D.; Barnes, L.L.; Mehta, R.I.; Dams-O’Connor, K.; Mez, J.; Bennett, D.A.; Schneider, J.A. Association of traumatic brain injury with and without loss of consciousness with neuropathologic outcomes in community-dwelling older persons. JAMA Netw. Open 2022, 5, e229311. [Google Scholar] [CrossRef]
  10. Crane, P.K.; Gibbons, L.E.; Dams-O’Connor, K.; Trittschuh, E.; Leverenz, J.B.; Keene, C.D.; Sonnen, J.; Montine, T.J.; Bennett, D.A.; Leurgans, S.; et al. Association of traumatic brain injury with late-life neurodegenerative conditions and neuropathologic findings. JAMA Neurol. 2016, 73, 1062–1069. [Google Scholar] [CrossRef]
  11. Gardner, R.C.; Bahorik, A.; Kornblith, E.S.; Allen, I.E.; Plassman, B.L.; Yaffe, K. Systematic Review, Meta-Analysis, and Population Attributable Risk of Dementia Associated with Traumatic Brain Injury in Civilians and Veterans. J. Neurotrauma 2023, 40, 620–634. [Google Scholar] [CrossRef] [PubMed]
  12. Hanrahan, J.G.; Burford, C.; Nagappan, P.; Adegboyega, G.; Rajkumar, S.; Kolias, A.; Helmy, A.; Hutchinson, P.J. Is dementia more likely following traumatic brain injury? A systematic review. J. Neurol. 2023, 270, 3022–3051. [Google Scholar] [CrossRef] [PubMed]
  13. Iadecola, C. The pathobiology of vascular dementia. Neuron 2013, 80, 844–866. [Google Scholar] [CrossRef] [PubMed]
  14. Pluta, R.; Furmaga-Jabłońska, W.; Maciejewski, R.; Ułamek-Kozioł, M.; Jabłoński, M. Brain ischemia activates β- and γ-secretase cleavage of amyloid precursor protein: Significance in sporadic Alzheimer’s disease. Mol. Neurobiol. 2013, 47, 425–434. [Google Scholar] [CrossRef] [PubMed]
  15. Ramos-Cejudo, J.; Wisniewski, T.; Marmar, C.; Zetterberg, H.; Blennow, K.; de Leon, M.J.; Fossati, S. Traumatic Brain Injury and Alzheimer’s Disease: The Cerebrovascular Link. EBioMedicine 2018, 28, 21–30. [Google Scholar] [CrossRef] [PubMed]
  16. Yuh, E.L.; Mukherjee, P.; Lingsma, H.F.; Yue, J.K.; Ferguson, A.R.; Gordon, W.A.; Valadka, A.B.; Schnyer, D.M.; Okonkwo, D.O.; Maas, A.I.; et al. Magnetic resonance imaging improves 3-month outcome prediction in mild traumatic brain injury. Ann. Neurol. 2013, 73, 224–235. [Google Scholar] [CrossRef] [PubMed]
  17. Kenney, K.; Amyot, F.; Haber, M.; Pronger, A.; Bogoslovsky, T.; Moore, C.; Diaz-Arrastia, R. Cerebral vascular injury in traumatic brain injury. Exp. Neurol. 2016, 275, 353–366. [Google Scholar] [CrossRef]
  18. Ichkova, A.; Rodriguez-Grande, B.; Bar, C.; Villega, F.; Konsman, J.P.; Badaut, J. Vascular impairment as a pathological mechanism underlying long-lasting cognitive dysfunction after pediatric traumatic brain injury. Neurochem. Int. 2017, 111, 93–102. [Google Scholar] [CrossRef] [PubMed]
  19. Ng, S.Y.; Lee, A.Y.W. Traumatic brain injuries: Pathophysiology and potential therapeutic targets. Front. Cell. Neurosci. 2019, 13, 528. [Google Scholar] [CrossRef] [PubMed]
  20. Harting, M.T.; Jimenez, F.; Adams, S.D.; Mercer, D.W.; Cox, C.S., Jr. Acute, regional inflammatory response after traumatic brain injury: Implications for cellular therapy. Surgery 2008, 144, 803–813. [Google Scholar] [CrossRef] [PubMed]
  21. Werner, C.; Engelhard, K. Pathophysiology of traumatic brain injury. Br. J. Anaesth. 2007, 99, 4–9. [Google Scholar] [CrossRef]
  22. Gaetz, M. The neurophysiology of brain injury. Clin. Neurophysiol. 2004, 115, 4–18. [Google Scholar] [CrossRef] [PubMed]
  23. Johnstone, V.P.; Shultz, S.R.; Yan, E.B.; O’Brien, T.J.; Rajan, R. The acute phase of mild traumatic brain injury is characterized by a distance-dependent neuronal hypoactivity. J. Neurotrauma 2014, 31, 1881–1895. [Google Scholar] [CrossRef] [PubMed]
  24. Hetz, C.; Saxena, S. ER stress and the unfolded protein response in neurodegeneration. Nat. Rev. Neurol. 2017, 13, 477–491. [Google Scholar] [CrossRef] [PubMed]
  25. Freire, M. Pathophysiology of neurodegeneration following traumatic brain injury. West Indian Med. J. 2012, 61, 751–755. [Google Scholar]
  26. Cruz-Haces, M.; Tang, J.; Acosta, G.; Fernandez, J.; Shi, R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl. Neurodegener. 2017, 6, 20. [Google Scholar] [CrossRef]
  27. Head, B.P.; Peart, J.N.; Panneerselvam, M.; Yokoyama, T.; Pearn, M.L.; Niesman, I.R.; Bonds, J.A.; Schilling, J.M.; Miyanohara, A.; Headrick, J.; et al. Loss of caveolin-1 accelerates neurodegeneration and aging. PLoS ONE 2010, 5, e15697. [Google Scholar] [CrossRef]
  28. Pearn, M.L.; Niesman, I.R.; Egawa, J.; Sawada, A.; Almenar-Queralt, A.; Shah, S.B.; Duckworth, J.L.; Head, B.P. Pathophysiology associated with traumatic brain injury: Current treatments and potential novel therapeutics. Cell. Mol. Neurobiol. 2017, 37, 571–585. [Google Scholar] [CrossRef] [PubMed]
  29. Shohami, E.; Beit-Yannai, E.; Horowitz, M.; Kohen, R. Oxidative stress in closed-head injury: Brain antioxidant capacity as an indicator of functional outcome. J. Cereb. Blood Flow Metab. 1997, 17, 1007–1019. [Google Scholar] [CrossRef] [PubMed]
  30. Massaad, C.A.; Klann, E. Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid. Redox Signal. 2011, 14, 2013–2054. [Google Scholar] [CrossRef] [PubMed]
  31. 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] [PubMed]
  32. Winkler, E.A.; Minter, D.; Yue, J.K.; Manley, G.T. Cerebral edema in traumatic brain injury: Pathophysiology and prospective therapeutic targets. Neurosurg. Clin. 2016, 27, 473–488. [Google Scholar] [CrossRef] [PubMed]
  33. Maragakis, N.J.; Rothstein, J.D. Glutamate transporters in neurologic disease. Arch. Neurol. 2001, 58, 365–370. [Google Scholar] [CrossRef]
  34. Bullock, R.; Zauner, A.; Woodward, J.J.; Myseros, J.; Choi, S.C.; Ward, J.D.; Marmarou, A.; Young, H.F. Factors affecting excitatory amino acid release following severe human head injury. J. Neurosurg. 1998, 89, 507–518. [Google Scholar] [CrossRef] [PubMed]
  35. Hlatky, R.; Valadka, A.B.; Goodman, J.C.; Contant, C.F.; Robertson, C.S. Patterns of energy substrates during ischemia measured in the brain by microdialysis. J. Neurotrauma 2004, 21, 894–906. [Google Scholar] [CrossRef] [PubMed]
  36. Sarrafzadeh, A.S.; Kiening, K.L.; Callsen, T.A.; Unterberg, A.W. Metabolic changes during impending and manifest cerebral hypoxia in traumatic brain injury. Br. J. Neurosurg. 2003, 17, 340–346. [Google Scholar] [CrossRef]
  37. Vespa, P.; Prins, M.; Ronne-Engstrom, E.; Caron, M.; Shalmon, E.; Hovda, D.A.; Martin, N.A.; Becker, D.P. Increase in extracellular glutamate caused by reduced cerebral perfusion pressure and seizures after human traumatic brain injury: A microdialysis study. J. Neurosurg. 1998, 89, 971–982. [Google Scholar] [CrossRef] [PubMed]
  38. Yamamoto, T.; Rossi, S.; Stiefel, M.; Doppenberg, E.; Zauner, A.; Bullock, R.; Marmarou, A. CSF and ECF glutamate concentrations in head injured patients. Acta Neurochir. Suppl. 1999, 75, 17–19. [Google Scholar]
  39. Rao, V.L.; Başkaya, M.K.; Doğan, A.; Rothstein, J.D.; Dempsey, R.J. Traumatic brain injury down-regulates glial glutamate transporter (GLT-1 and GLAST) proteins in rat brain. J. Neurochem. 1998, 70, 2020–2027. [Google Scholar]
  40. Van Landeghem, F.K.; Stover, J.F.; Bechmann, I.; Brück, W.; Unterberg, A.; Bührer, C.; von Deimling, A. Early expression of glutamate transporter proteins in ramified microglia after controlled cortical impact injury in the rat. Glia 2001, 35, 167–179. [Google Scholar] [CrossRef]
  41. Yi, J.H.; Hazell, A.S. Excitotoxic mechanisms and the role of astrocytic glutamate transporters in traumatic brain injury. Neurochem. Int. 2006, 48, 394–403. [Google Scholar] [CrossRef] [PubMed]
  42. Huang, F.J.; You, W.K.; Bonaldo, P.; Seyfried, T.N.; Pasquale, E.B.; Stallcup, W.B. Pericyte deficiencies lead to aberrant tumor vascularizaton in the brain of the NG2 null mouse. Dev. Biol. 2010, 344, 1035–1046. [Google Scholar] [CrossRef] [PubMed]
  43. Rothstein, J.D.; Dykes-Hoberg, M.; Pardo, C.A.; Bristol, L.A.; Jin, L.; Kuncl, R.W.; Kanai, Y.; Hediger, M.A.; Wang, Y.; Schielke, J.P.; et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron 1996, 16, 675–686. [Google Scholar] [CrossRef]
  44. Van Landeghem, F.K.; Weiss, T.; Oehmichen, M.; von Deimling, A. Decreased expression of glutamate transporters in astrocytes after human traumatic brain injury. J. Neurotrauma 2006, 23, 1518–1528. [Google Scholar] [CrossRef] [PubMed]
  45. Barres, B.A. The mystery and magic of glia: A perspective on their roles in health and disease. Neuron 2008, 60, 430–440. [Google Scholar] [CrossRef] [PubMed]
  46. Rogers, S.D.; Peters, C.M.; Pomonis, J.D.; Hagiwara, H.; Ghilardi, J.R.; Mantyh, P.W. Endothelin B receptors are expressed by astrocytes and regulate astrocyte hypertrophy in the normal and injured CNS. Glia 2003, 41, 180–190. [Google Scholar] [CrossRef]
  47. Van Gent, J.A.; van Essen, T.A.; Bos, M.H.; Cannegieter, S.C.; van Dijck, J.T.; Peul, W.C. Coagulopathy after hemorrhagic traumatic brain injury, an observational study of the incidence and prognosis. Acta Neurochir. 2020, 162, 329–336. [Google Scholar] [CrossRef]
  48. Engström, M.; Romner, B.; Schalén, W.; Reinstrup, P. Thrombocytopenia predicts progressive hemorrhage after head trauma. J. Neurotrauma 2005, 22, 291–296. [Google Scholar] [CrossRef]
  49. Lustenberger, T.; Talving, P.; Kobayashi, L.; Barmparas, G.; Inaba, K.; Lam, L.; Branco, B.C.; Demetriades, D. Early coagulopathy after isolated severe traumatic brain injury: Relationship with hypoperfusion challenged. J. Trauma Acute Care Surg. 2010, 69, 1410–1414. [Google Scholar] [CrossRef]
  50. Cohen, M.J.; Brohi, K.; Ganter, M.T.; Manley, G.T.; Mackersie, R.C.; Pittet, J.-F. Early coagulopathy after traumatic brain injury: The role of hypoperfusion and the protein C pathway. J. Trauma Acute Care Surg. 2007, 63, 1254–1262. [Google Scholar] [CrossRef]
  51. Greuters, S.; van den Berg, A.; Franschman, G.; Viersen, V.A.; Beishuizen, A.; Peerdeman, S.M.; Boer, C. Acute and delayed mild coagulopathy are related to outcome in patients with isolated traumatic brain injury. Crit. Care 2011, 15, R2. [Google Scholar] [CrossRef] [PubMed]
  52. Oertel, M.; Kelly, D.F.; McArthur, D.; Boscardin, W.J.; Glenn, T.C.; Lee, J.H.; Gravori, T.; Obukhov, D.; McBride, D.Q.; Martin, N.A. Progressive hemorrhage after head trauma: Predictors and consequences of the evolving injury. J. Neurosurg. 2002, 96, 109–116. [Google Scholar] [CrossRef] [PubMed]
  53. Irimia, A.; Chambers, M.C.; Alger, J.R.; Filippou, M.; Prastawa, M.W.; Wang, B.; Hovda, D.A.; Gerig, G.; Toga, A.W.; Kikinis, R.; et al. Comparison of acute and chronic traumatic brain injury using semi-automatic multimodal segmentation of MR volumes. J. Neurotrauma 2011, 28, 2287–2306. [Google Scholar] [CrossRef]
  54. Iwamura, A.; Taoka, T.; Fukusumi, A.; Sakamoto, M.; Miyasaka, T.; Ochi, T.; Akashi, T.; Okuchi, K.; Kichikawa, K. Diffuse vascular injury: Convergent-type hemorrhage in the supratentorial white matter on susceptibility-weighted image in cases of severe traumatic brain damage. Neuroradiology 2012, 54, 335–343. [Google Scholar] [CrossRef] [PubMed]
  55. Kinnunen, K.M.; Greenwood, R.; Powell, J.H.; Leech, R.; Hawkins, P.C.; Bonnelle, V.; Patel, M.C.; Counsell, S.J.; Sharp, D.J. White matter damage and cognitive impairment after traumatic brain injury. Brain 2011, 134 Pt 2, 449–463. [Google Scholar] [CrossRef]
  56. Gaasch, J.A.; Lockman, P.R.; Geldenhuys, W.J.; Allen, D.D.; Van der Schyf, C.J. Brain iron toxicity: Differential responses of astrocytes, neurons, and endothelial cells. Neurochem. Res. 2007, 32, 1196–1208. [Google Scholar] [CrossRef]
  57. Lok, J.; Leung, W.; Murphy, S.; Butler, W.; Noviski, N.; Lo, E.H. Intracranial hemorrhage: Mechanisms of secondary brain injury. Acta Neurochir. Suppl. 2011, 111, 63–69. [Google Scholar]
  58. Schrag, M.; McAuley, G.; Pomakian, J.; Jiffry, A.; Tung, S.; Mueller, C.; Vinters, H.V.; Haacke, E.M.; Holshouser, B.; Kido, D.; et al. Correlation of hypointensities in susceptibility-weighted images to tissue histology in dementia patients with cerebral amyloid angiopathy: A postmortem MRI study. Acta Neuropathol. 2010, 119, 291–302. [Google Scholar] [CrossRef]
  59. Johnson, V.E.; Stewart, J.E.; Begbie, F.D.; Trojanowski, J.Q.; Smith, D.H.; Stewart, W. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain 2013, 136 Pt 1, 28–42. [Google Scholar] [CrossRef]
  60. Fujita, M.; Wei, E.P.; Povlishock, J.T. Intensity- and interval-specific repetitive traumatic brain injury can evoke both axonal and microvascular damage. J. Neurotrauma 2012, 29, 2172–2180. [Google Scholar] [CrossRef]
  61. Lin, B.; Ginsberg, M.D.; Zhao, W.; Alonso, O.F.; Belayev, L.; Busto, R. Quantitative analysis of microvascular alterations in traumatic brain injury by endothelial barrier antigen immunohistochemistry. J. Neurotrauma 2001, 18, 389–397. [Google Scholar] [CrossRef] [PubMed]
  62. Sangiorgi, S.; De Benedictis, A.; Protasoni, M.; Manelli, A.; Reguzzoni, M.; Cividini, A.; Dell’orbo, C.; Tomei, G.; Balbi, S. Early-stage microvascular alterations of a new model of controlled cortical traumatic brain injury: 3D morphological analysis using scanning electron microscopy and corrosion casting. J. Neurosurg. 2013, 118, 763–774. [Google Scholar] [CrossRef] [PubMed]
  63. Wei, E.P.; Hamm, R.J.; Baranova, A.I.; Povlishock, J.T. The long-term microvascular and behavioral consequences of experimental traumatic brain injury after hypothermic intervention. J. Neurotrauma 2009, 26, 527–537. [Google Scholar] [CrossRef]
  64. Li, C.; Wang, Y.; Yan, X.L.; Guo, Z.N.; Yang, Y. Pathological changes in neurovascular units: Lessons from cases of vascular dementia. CNS Neurosci. Ther. 2021, 27, 17–25. [Google Scholar] [CrossRef] [PubMed]
  65. Iadecola, C. The neurovascular unit coming of age: A journey through neurovascular coupling in health and disease. Neuron 2017, 96, 17–42. [Google Scholar] [CrossRef] [PubMed]
  66. Jessen, K.R. Glial cells. Int. J. Biochem. Cell Biol. 2004, 36, 1861–1867. [Google Scholar] [CrossRef]
  67. Sweeney, M.D.; Ayyadurai, S.; Zlokovic, B.V. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat. Neurosci. 2016, 19, 771–783. [Google Scholar] [CrossRef]
  68. Stanimirovic, D.B.; Friedman, A. Pathophysiology of the neurovascular unit: Disease cause or consequence? J. Cereb. Blood Flow Metab. 2012, 32, 1207–1221. [Google Scholar] [CrossRef]
  69. Del Zoppo, G.J. The neurovascular unit, matrix proteases, and innate inflammation. Ann. N. Y. Acad. Sci. 2010, 1207, 46–49. [Google Scholar] [CrossRef]
  70. ElAli, A. Neurovascular unit dysfunction in dementia: A brief summary. Austin Alzheimer’s Park. Dis. 2014, 1, 5. [Google Scholar]
  71. Jullienne, A.; Obenaus, A.; Ichkova, A.; Savona-Baron, C.; Pearce, W.J.; Badaut, J. Chronic cerebrovascular dysfunction after traumatic brain injury. J. Neurosci. Res. 2016, 94, 609–622. [Google Scholar] [CrossRef] [PubMed]
  72. Charkviani, M.; Muradashvili, N.; Lominadze, D. Vascular and non-vascular contributors to memory reduction during traumatic brain injury. Eur. J. Neurosci. 2019, 50, 2860–2876. [Google Scholar] [CrossRef] [PubMed]
  73. Hay, J.R.; Johnson, V.E.; Young, A.M.; Smith, D.H.; Stewart, W. Blood-brain barrier disruption is an early event that may persist for many years after traumatic brain injury in humans. J. Neuropathol. Exp. Neurol. 2015, 74, 1147–1157. [Google Scholar] [PubMed]
  74. Hellström, M.; Kalén, M.; Lindahl, P.; Abramsson, A.; Betsholtz, C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 1999, 126, 3047–3055. [Google Scholar] [CrossRef]
  75. Lindahl, P.; Johansson, B.R.; Levéen, P.; Betsholtz, C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science 1997, 277, 242–245. [Google Scholar] [CrossRef]
  76. Abdul-Muneer, P.M.; Bhowmick, S.; Briski, N. Angiotensin II Causes Neuronal Damage in Stretch-Injured Neurons: Protective Effects of Losartan, an Angiotensin T(1) Receptor Blocker. Mol. Neurobiol. 2018, 55, 5901–5912. [Google Scholar] [CrossRef]
  77. Abdul-Muneer, P.M.; Long, M.; Conte, A.A.; Santhakumar, V.; Pfister, B.J. High Ca(2+) Influx During Traumatic Brain Injury Leads to Caspase-1-Dependent Neuroinflammation and Cell Death. Mol. Neurobiol. 2017, 54, 3964–3975. [Google Scholar] [CrossRef]
  78. Takata, F.; Dohgu, S.; Matsumoto, J.; Takahashi, H.; Machida, T.; Wakigawa, T.; Harada, E.; Miyaji, H.; Koga, M.; Nishioku, T.; et al. Brain pericytes among cells constituting the blood-brain barrier are highly sensitive to tumor necrosis factor-α, releasing matrix metalloproteinase-9 and migrating in vitro. J. Neuroinflammation 2011, 8, 106. [Google Scholar] [CrossRef]
  79. Kunz, J.; Krause, D.; Kremer, M.; Dermietzel, R. The 140-kDa protein of blood-brain barrier-associated pericytes is identical to aminopeptidase N. J. Neurochem. 1994, 62, 2375–2386. [Google Scholar] [CrossRef]
  80. Winkler, E.A.; Bell, R.D.; Zlokovic, B.V. Pericyte-specific expression of PDGF beta receptor in mouse models with normal and deficient PDGF beta receptor signaling. Mol. Neurodegener. 2010, 5, 32. [Google Scholar] [CrossRef]
  81. Armulik, A.; Genové, G.; Betsholtz, C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell 2011, 21, 193–215. [Google Scholar] [CrossRef]
  82. Bhowmick, S.; D’Mello, V.; Caruso, D.; Wallerstein, A.; Abdul-Muneer, P.M. Impairment of pericyte-endothelium crosstalk leads to blood-brain barrier dysfunction following traumatic brain injury. Exp. Neurol. 2019, 317, 260–270. [Google Scholar] [CrossRef] [PubMed]
  83. Bjarnegård, M.; Enge, M.; Norlin, J.; Gustafsdottir, S.; Fredriksson, S.; Abramsson, A.; Takemoto, M.; Gustafsson, E.; Fässler, R.; Betsholtz, C. Endothelium-specific ablation of PDGFB leads to pericyte loss and glomerular, cardiac and placental abnormalities. Development 2004, 131, 1847–1857. [Google Scholar] [CrossRef]
  84. Enge, M.; Bjarnegård, M.; Gerhardt, H.; Gustafsson, E.; Kalén, M.; Asker, N.; Hammes, H.P.; Shani, M.; Fässler, R.; Betsholtz, C. Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy. Embo J. 2002, 21, 4307–4316. [Google Scholar] [CrossRef] [PubMed]
  85. Armulik, A.; Abramsson, A.; Betsholtz, C. Endothelial/pericyte interactions. Circ. Res. 2005, 97, 512–523. [Google Scholar] [CrossRef]
  86. Lebrin, F.; Srun, S.; Raymond, K.; Martin, S.; van den Brink, S.; Freitas, C.; Bréant, C.; Mathivet, T.; Larrivée, B.; Thomas, J.L.; et al. Thalidomide stimulates vessel maturation and reduces epistaxis in individuals with hereditary hemorrhagic telangiectasia. Nat. Med. 2010, 16, 420–428. [Google Scholar] [CrossRef] [PubMed]
  87. Li, F.; Lu, L.; Shang, S.; Chen, H.; Wang, P.; Haidari, N.A.; Chen, Y.C.; Yin, X. Cerebral blood flow and its connectivity deficits in mild traumatic brain injury at the acute stage. Neural Plast. 2020, 2020, 2174371. [Google Scholar] [CrossRef] [PubMed]
  88. Inoue, Y.; Shiozaki, T.; Tasaki, O.; Hayakata, T.; Ikegawa, H.; Yoshiya, K.; Fujinaka, T.; Tanaka, H.; Shimazu, T.; Sugimoto, H. Changes in cerebral blood flow from the acute to the chronic phase of severe head injury. J. Neurotrauma 2005, 22, 1411–1418. [Google Scholar] [CrossRef]
  89. Washington, P.M.; Lee, C.; Dwyer, M.K.R.; Konofagou, E.E.; Kernie, S.G.; Morrison, I.I.I.B. Hyaluronidase reduced edema after experimental traumatic brain injury. J. Cereb. Blood Flow Metab. 2020, 40, 2026–2037. [Google Scholar] [CrossRef]
  90. Balestreri, M.; Czosnyka, M.; Hutchinson, P.; Steiner, L.A.; Hiler, M.; Smielewski, P.; Pickard, J.D. Impact of intracranial pressure and cerebral perfusion pressure on severe disability and mortality after head injury. Neurocritical Care 2006, 4, 8–13. [Google Scholar] [CrossRef]
  91. Marmarou, A.; Fatouros, P.P.; Barzó, P.; Portella, G.; Yoshihara, M.; Tsuji, O.; Yamamoto, T.; Laine, F.; Signoretti, S.; Ward, J.D.; et al. Contribution of edema and cerebral blood volume to traumatic brain swelling in head-injured patients. J. Neurosurg. 2000, 93, 183–193. [Google Scholar] [CrossRef] [PubMed]
  92. Dewan, M.C.; Rattani, A.; Gupta, S.; Baticulon, R.E.; Hung, Y.C.; Punchak, M.; Agrawal, A.; Adeleye, A.O.; Shrime, M.G.; Rubiano, A.M.; et al. Estimating the global incidence of traumatic brain injury. J. Neurosurg. 2018, 130, 1080–1097. [Google Scholar] [CrossRef] [PubMed]
  93. Das, M.; Mohapatra, S.; Mohapatra, S.S. New perspectives on central and peripheral immune responses to acute traumatic brain injury. J. Neuroinflammation 2012, 9, 236. [Google Scholar] [CrossRef]
  94. Faden, A.I. Microglial activation and traumatic brain injury. Ann. Neurol. 2011, 70, 345–346. [Google Scholar] [CrossRef]
  95. Hsieh, C.L.; Kim, C.C.; Ryba, B.E.; Niemi, E.C.; Bando, J.K.; Locksley, R.M.; Liu, J.; Nakamura, M.C.; Seaman, W.E. Traumatic brain injury induces macrophage subsets in the brain. Eur. J. Immunol. 2013, 43, 2010–2022. [Google Scholar] [CrossRef] [PubMed]
  96. Ramlackhansingh, A.F.; Brooks, D.J.; Greenwood, R.J.; Bose, S.K.; Turkheimer, F.E.; Kinnunen, K.M.; Gentleman, S.; Heckemann, R.A.; Gunanayagam, K.; Gelosa, G.; et al. Inflammation after trauma: Microglial activation and traumatic brain injury. Ann. Neurol. 2011, 70, 374–383. [Google Scholar] [CrossRef]
  97. Witcher, K.G.; Eiferman, D.S.; Godbout, J.P. Priming the inflammatory pump of the CNS after traumatic brain injury. Trends Neurosci. 2015, 38, 609–620. [Google Scholar] [CrossRef]
  98. Gao, C.; Qian, Y.; Huang, J.; Wang, D.; Su, W.; Wang, P.; Guo, L.; Quan, W.; An, S.; Zhang, J.; et al. A Three-Day Consecutive Fingolimod Administration Improves Neurological Functions and Modulates Multiple Immune Responses of CCI Mice. Mol. Neurobiol. 2017, 54, 8348–8360. [Google Scholar] [CrossRef]
  99. Gyoneva, S.; Ransohoff, R.M. Inflammatory reaction after traumatic brain injury: Therapeutic potential of targeting cell-cell communication by chemokines. Trends Pharmacol. Sci. 2015, 36, 471–480. [Google Scholar] [CrossRef]
  100. Xu, X.; Gao, W.; Cheng, S.; Yin, D.; Li, F.; Wu, Y.; Sun, D.; Zhou, S.; Wang, D.; Zhang, Y.; et al. Anti-inflammatory and immunomodulatory mechanisms of atorvastatin in a murine model of traumatic brain injury. J. Neuroinflamm. 2017, 14, 167. [Google Scholar] [CrossRef]
  101. Amor, S.; Puentes, F.; Baker, D.; van der Valk, P. Inflammation in neurodegenerative diseases. Immunology 2010, 129, 154–169. [Google Scholar] [CrossRef] [PubMed]
  102. 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] [PubMed]
  103. Turrin, N.P.; Plante, M.-M.; Lessard, M.; Rivest, S. Irradiation does not compromise or exacerbate the innate immune response in the brains of mice that were transplanted with bone marrow stem cells. Stem Cells 2007, 25, 3165–3172. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, G.; Zhang, J.; Hu, X.; Zhang, L.; Mao, L.; Jiang, X.; Liou, A.K.; Leak, R.K.; Gao, Y.; Chen, J. Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J. Cereb. Blood Flow Metab. 2013, 33, 1864–1874. [Google Scholar] [CrossRef]
  105. Ritzel, R.M.; Doran, S.J.; Barrett, J.P.; Henry, R.J.; Ma, E.L.; Faden, A.I.; Loane, D.J. Chronic Alterations in Systemic Immune Function after Traumatic Brain Injury. J. Neurotrauma 2018, 35, 1419–1436. [Google Scholar] [CrossRef]
  106. Simon, D.W.; McGeachy, M.J.; Bayır, H.; Clark, R.S.; 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]
  107. Sofroniew, M.V. Astrocyte barriers to neurotoxic inflammation. Nat. Rev. Neurosci. 2015, 16, 249–263. [Google Scholar] [CrossRef]
  108. Coughlin, J.M.; Wang, Y.; Munro, C.A.; Ma, S.; Yue, C.; Chen, S.; Airan, R.; Kim, P.K.; Adams, A.V.; Garcia, C.; et al. Neuroinflammation and brain atrophy in former NFL players: An in vivo multimodal imaging pilot study. Neurobiol. Dis. 2015, 74, 58–65. [Google Scholar] [CrossRef]
  109. Gentleman, S.M.; Leclercq, P.D.; Moyes, L.; Graham, D.I.; Smith, C.; Griffin, W.S.; Nicoll, J.A. Long-term intracerebral inflammatory response after traumatic brain injury. Forensic. Sci. Int. 2004, 146, 97–104. [Google Scholar] [CrossRef]
  110. 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]
  111. Muccigrosso, M.M.; Ford, J.; Benner, B.; Moussa, D.; Burnsides, C.; Fenn, A.M.; Popovich, P.G.; Lifshitz, J.; Walker, F.R.; Eiferman, D.S.; et al. Cognitive deficits develop 1month after diffuse brain injury and are exaggerated by microglia-associated reactivity to peripheral immune challenge. Brain Behav. Immun. 2016, 54, 95–109. [Google Scholar] [CrossRef]
  112. Hu, X.; Li, P.; Guo, Y.; Wang, H.; Leak, R.K.; Chen, S.; Gao, Y.; Chen, J. Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke 2012, 43, 3063–3070. [Google Scholar] [CrossRef] [PubMed]
  113. Kigerl, K.A.; Gensel, J.C.; Ankeny, D.P.; Alexander, J.K.; Donnelly, D.J.; Popovich, P.G. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J. Neurosci. 2009, 29, 13435–13444. [Google Scholar] [CrossRef] [PubMed]
  114. Guo, X.; Liu, L.; Zhang, M.; Bergeron, A.; Cui, Z.; Dong, J.F.; Zhang, J. Correlation of CD34+ cells with tissue angiogenesis after traumatic brain injury in a rat model. J. Neurotrauma 2009, 26, 1337–1344. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, Z.G.; Zhang, L.; Jiang, Q.; Chopp, M. Bone marrow-derived endothelial progenitor cells participate in cerebral neovascularization after focal cerebral ischemia in the adult mouse. Circ. Res. 2002, 90, 284–288. [Google Scholar] [CrossRef]
  116. Dvorak, H.F.; Brown, L.F.; Detmar, M.; Dvorak, A.M. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am. J. Pathol. 1995, 146, 1029–1039. [Google Scholar]
  117. Sköld, M.K.; von Gertten, C.; Sandberg-Nordqvist, A.C.; Mathiesen, T.; Holmin, S. VEGF and VEGF receptor expression after experimental brain contusion in rat. J. Neurotrauma 2005, 22, 353–367. [Google Scholar] [CrossRef]
  118. Mascia, L.; Sakr, Y.; Pasero, D.; Payen, D.; Reinhart, K.; Vincent, J.L. Extracranial complications in patients with acute brain injury: A post-hoc analysis of the SOAP study. Intensive Care Med. 2008, 34, 720–727. [Google Scholar] [CrossRef]
  119. Jeremitsky, E.; Omert, L.; Dunham, C.M.; Protetch, J.; Rodriguez, A. Harbingers of poor outcome the day after severe brain injury: Hypothermia, hypoxia, and hypoperfusion. J. Trauma 2003, 54, 312–319. [Google Scholar] [CrossRef]
  120. Krishnamoorthy, V.; Komisarow, J.M.; Laskowitz, D.T.; Vavilala, M.S. Multiorgan Dysfunction After Severe Traumatic Brain Injury: Epidemiology, Mechanisms, and Clinical Management. Chest 2021, 160, 956–964. [Google Scholar] [CrossRef]
  121. Sharma, R.; Shultz, S.R.; Robinson, M.J.; Belli, A.; Hibbs, M.L.; O’Brien, T.J.; Semple, B.D. Infections after a traumatic brain injury: The complex interplay between the immune and neurological systems. Brain Behav. Immun. 2019, 79, 63–74. [Google Scholar] [CrossRef]
  122. Krishnamoorthy, V.; Rowhani-Rahbar, A.; Gibbons, E.F.; Rivara, F.P.; Temkin, N.R.; Pontius, C.; Luk, K.; Graves, M.; Lozier, D.; Chaikittisilpa, N. Early Systolic Dysfunction Following Traumatic Brain Injury: A Cohort Study. Crit. Care Med. 2017, 45, 1028–1036. [Google Scholar] [CrossRef]
  123. Krishnamoorthy, V.; Mackensen, G.B.; Gibbons, E.F.; Vavilala, M.S. Cardiac Dysfunction After Neurologic Injury: What Do We Know and Where Are We Going? Chest 2016, 149, 1325–1331. [Google Scholar] [CrossRef]
  124. Krishnamoorthy, V.; Rowhani-Rahbar, A.; Chaikittisilpa, N.; Gibbons, E.F.; Rivara, F.P.; Temkin, N.R.; Quistberg, A.; Vavilala, M.S. Association of Early Hemodynamic Profile and the Development of Systolic Dysfunction Following Traumatic Brain Injury. Neurocrit. Care 2017, 26, 379–387. [Google Scholar] [CrossRef] [PubMed]
  125. Samuels, M.A. The brain-heart connection. Circulation 2007, 116, 77–84. [Google Scholar] [CrossRef] [PubMed]
  126. Wafaisade, A.; Lefering, R.; Tjardes, T.; Wutzler, S.; Simanski, C.; Paffrath, T.; Fischer, P.; Bouillon, B.; Maegele, M. Acute coagulopathy in isolated blunt traumatic brain injury. Neurocrit. Care 2010, 12, 211–219. [Google Scholar] [CrossRef]
  127. Stein, S.C.; Young, G.S.; Talucci, R.C.; Greenbaum, B.H.; Ross, S.E. Delayed brain injury after head trauma: Significance of coagulopathy. Neurosurgery 1992, 30, 160–165. [Google Scholar] [CrossRef] [PubMed]
  128. Maegele, M. Coagulopathy after traumatic brain injury: Incidence, pathogenesis, and treatment options. Transfusion 2013, 53 (Suppl. 1), 28s–37s. [Google Scholar] [CrossRef]
  129. Harhangi, B.S.; Kompanje, E.J.; Leebeek, F.W.; Maas, A.I. Coagulation disorders after traumatic brain injury. Acta Neurochir. 2008, 150, 165–175, discussion 175. [Google Scholar] [CrossRef]
  130. Epstein, D.S.; Mitra, B.; O’Reilly, G.; Rosenfeld, J.V.; Cameron, P.A. Acute traumatic coagulopathy in the setting of isolated traumatic brain injury: A systematic review and meta-analysis. Injury 2014, 45, 819–824. [Google Scholar] [CrossRef]
  131. Laroche, M.; Kutcher, M.E.; Huang, M.C.; Cohen, M.J.; Manley, G.T. Coagulopathy after traumatic brain injury. Neurosurgery 2012, 70, 1334–1345. [Google Scholar] [CrossRef]
  132. Gupte, R.; Brooks, W.; Vukas, R.; Pierce, J.; Harris, J. Sex Differences in Traumatic Brain Injury: What We Know and What We Should Know. J. Neurotrauma 2019, 36, 3063–3091. [Google Scholar] [CrossRef] [PubMed]
  133. Chang, V.C.; Ruseckaite, R.; Collie, A.; Colantonio, A. Examining the epidemiology of work-related traumatic brain injury through a sex/gender lens: Analysis of workers’ compensation claims in Victoria, Australia. Occup. Environ. Med. 2014, 71, 695–703. [Google Scholar] [CrossRef] [PubMed]
  134. Colantonio, A. Sex, Gender, and Traumatic Brain Injury: A Commentary. Arch. Phys. Med. Rehabil. 2016, 97 (Suppl. 2), S1–S4. [Google Scholar] [CrossRef]
  135. Iverson, K.M.; Hendricks, A.M.; Kimerling, R.; Krengel, M.; Meterko, M.; Stolzmann, K.L.; Baker, E.; Pogoda, T.K.; Vasterling, J.J.; Lew, H.L. Psychiatric diagnoses and neurobehavioral symptom severity among OEF/OIF VA patients with deployment-related traumatic brain injury: A gender comparison. Womens Health Issues 2011, 21 (Suppl. 4), S210–S217. [Google Scholar] [CrossRef]
  136. Czosnyka, M.; Radolovich, D.; Balestreri, M.; Lavinio, A.; Hutchinson, P.; Timofeev, I.; Smielewski, P.; Pickard, J.D. Gender-related differences in intracranial hypertension and outcome after traumatic brain injury. Acta Neurochir. Suppl. 2008, 102, 25–28. [Google Scholar] [PubMed]
  137. Whitehead, B.; Velazquez-Cruz, R.; Albowaidey, A.; Zhang, N.; Karelina, K.; Weil, Z.M. Mild Traumatic Brain Injury Induces Time- and Sex-Dependent Cerebrovascular Dysfunction and Stroke Vulnerability. J. Neurotrauma 2023, 40, 578–591. [Google Scholar] [CrossRef]
  138. Newell, E.A.; Todd, B.P.; Luo, Z.; Evans, L.P.; Ferguson, P.J.; Bassuk, A.G. A Mouse Model for Juvenile, Lateral Fluid Percussion Brain Injury Reveals Sex-Dependent Differences in Neuroinflammation and Functional Recovery. J. Neurotrauma. 2020, 37, 635–646. [Google Scholar] [CrossRef]
  139. O’Connor, C.A.; Cernak, I.; Vink, R. The temporal profile of edema formation differs between male and female rats following diffuse traumatic brain injury. Acta Neurochir. Suppl. 2006, 96, 121–124. [Google Scholar]
  140. Sayeed, I.; Wali, B.; Guthrie, D.B.; Saindane, M.T.; Natchus, M.G.; Liotta, D.C.; Stein, D.G. Development of a novel progesterone analog in the treatment of traumatic brain injury. Neuropharmacology 2019, 145, 292–298. [Google Scholar] [CrossRef]
  141. Xiao, G.; Wei, J.; Yan, W.; Wang, W.; Lu, Z. Improved outcomes from the administration of progesterone for patients with acute severe traumatic brain injury: A randomized controlled trial. Crit. Care 2008, 12, R61. [Google Scholar] [CrossRef] [PubMed]
  142. Brotfain, E.; Gruenbaum, S.E.; Boyko, M.; Kutz, R.; Zlotnik, A.; Klein, M. Neuroprotection by Estrogen and Progesterone in Traumatic Brain Injury and Spinal Cord Injury. Curr. Neuropharmacol. 2016, 14, 641–653. [Google Scholar] [CrossRef] [PubMed]
  143. Ludwig, P.E.; Patil, A.A.; Chamczuk, A.J.; Agrawal, D.K. Hormonal therapy in traumatic spinal cord injury. Am. J. Transl. Res. 2017, 9, 3881–3895. [Google Scholar]
  144. Bramlett, H.M.; Dietrich, W.D. Neuropathological protection after traumatic brain injury in intact female rats versus males or ovariectomized females. J. Neurotrauma 2001, 18, 891–900. [Google Scholar] [CrossRef] [PubMed]
  145. Giacometti, L.L.; Huh, J.W.; Raghupathi, R. Sex and estrous-phase dependent alterations in depression-like behavior following mild traumatic brain injury in adolescent rats. J. Neurosci. Res. 2022, 100, 490–505. [Google Scholar] [CrossRef] [PubMed]
  146. Krishna, G.; Bromberg, C.; Connell, E.C.; Mian, E.; Hu, C.; Lifshitz, J.; Adelson, P.D.; Thomas, T.C. Traumatic brain injury-induced sex-dependent changes in late-onset sensory hypersensitivity and glutamate neurotransmission. Front. Neurol. 2020, 11, 749. [Google Scholar] [CrossRef]
  147. Fortress, A.M.; Avcu, P.; Wagner, A.K.; Dixon, C.E.; Pang, K.C.H. Experimental traumatic brain injury results in estrous cycle disruption, neurobehavioral deficits, and impaired GSK3β/β-catenin signaling in female rats. Exp. Neurol. 2019, 315, 42–51. [Google Scholar] [CrossRef]
  148. Farace, E.; Alves, W.M. Do women fare worse: A metaanalysis of gender differences in traumatic brain injury outcome. J. Neurosurg. 2000, 93, 539–545. [Google Scholar] [CrossRef] [PubMed]
  149. Berry, C.; Ley, E.J.; Tillou, A.; Cryer, G.; Margulies, D.R.; Salim, A. The effect of gender on patients with moderate to severe head injuries. J. Trauma 2009, 67, 950–953. [Google Scholar] [CrossRef]
  150. Davis, D.P.; Douglas, D.J.; Smith, W.; Sise, M.J.; Vilke, G.M.; Holbrook, T.L.; Kennedy, F.; Eastman, A.B.; Velky, T.; Hoyt, D.B. Traumatic brain injury outcomes in pre- and post- menopausal females versus age-matched males. J. Neurotrauma 2006, 23, 140–148. [Google Scholar] [CrossRef] [PubMed]
  151. Wunderle, K.; Hoeger, K.M.; Wasserman, E.; Bazarian, J.J. Menstrual phase as predictor of outcome after mild traumatic brain injury in women. J. Head Trauma Rehabil. 2014, 29, E1–E8. [Google Scholar] [CrossRef] [PubMed]
  152. Ott, S.; Redell, J.; Cheema, S.; Schatz, P.; Becker, E. Progesterone Levels in Adolescent Female Athletes May Contribute to Decreased Cognitive Performance During Acute Phase of Sports-Related Concussion. Dev. Neuropsychol. 2024, 49, 86–97. [Google Scholar] [CrossRef]
  153. Ripley, D.L.; Harrison-Felix, C.; Sendroy-Terrill, M.; Cusick, C.P.; Dannels-McClure, A.; Morey, C. The impact of female reproductive function on outcomes after traumatic brain injury. Arch. Phys. Med. Rehabil. 2008, 89, 1090–1096. [Google Scholar] [CrossRef] [PubMed]
  154. Agha, A.; Thompson, C.J. Anterior pituitary dysfunction following traumatic brain injury (TBI). Clin. Endocrinol. 2006, 64, 481–488. [Google Scholar] [CrossRef] [PubMed]
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

Hasanpour-Segherlou, Z.; Masheghati, F.; Shakeri-Darzehkanani, M.; Hosseini-Siyanaki, M.-R.; Lucke-Wold, B. Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury. J. Vasc. Dis. 2024, 3, 319-332. https://doi.org/10.3390/jvd3030025

AMA Style

Hasanpour-Segherlou Z, Masheghati F, Shakeri-Darzehkanani M, Hosseini-Siyanaki M-R, Lucke-Wold B. Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury. Journal of Vascular Diseases. 2024; 3(3):319-332. https://doi.org/10.3390/jvd3030025

Chicago/Turabian Style

Hasanpour-Segherlou, Zahra, Forough Masheghati, Mahdieh Shakeri-Darzehkanani, Mohammad-Reza Hosseini-Siyanaki, and Brandon Lucke-Wold. 2024. "Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury" Journal of Vascular Diseases 3, no. 3: 319-332. https://doi.org/10.3390/jvd3030025

APA Style

Hasanpour-Segherlou, Z., Masheghati, F., Shakeri-Darzehkanani, M., Hosseini-Siyanaki, M.-R., & Lucke-Wold, B. (2024). Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury. Journal of Vascular Diseases, 3(3), 319-332. https://doi.org/10.3390/jvd3030025

Article Metrics

Back to TopTop