ABSTRACT
Background
Traumatic brain injury (TBI) is a leading cause of death and disability across all age groups worldwide. After primary mechanical head injury, a cascade of molecular changes and immunological responses occur that are necessary for supporting tissue repair but also exacerbate the secondary loss of tissue caused by excessive neuroinflammation. To date, there are no targeted treatments that ameliorate the pathological neuroinflammation that is responsible for propagating secondary injury after TBI. Recent works have highlighted the adaptive immune system's response to TBI, with mounting evidence suggesting that T cells play a critical yet understudied role in propagating secondary injury while also potentially supporting reparative processes.
Objectives
We critically review the current literature to discuss the diverse functionality of T cells in TBI including the temporospatial characteristics of their response, mechanisms of their activation, and their contributions to the overall neuroinflammatory profile. Consideration is given for additional pathological factors that may further alter these properties. We additionally summarize previous reports of therapeutic T cell modulation in this setting and identify approaches warranting additional investigation. Finally, we discuss major gaps in the existing literature and recommend future research perspectives.
Conclusion
Evidence suggests several aspects of the T cell response to TBI may serve as beneficial therapeutic targets for limiting secondary injury. Additional translational investigations are warranted and may support the development of effective therapeutic strategies for treating patients post‐head trauma.
Keywords: acute brain injury, neuroinflammation, T cells, T lymphocytes, traumatic brain injury
Growing evidence suggests T cells play a vital role in shaping the neuroinflammatory milieu that develops following traumatic brain injury (TBI). Pharmacological approaches for therapeutically modulating the contributions made by T cells to the pathophysiology of TBI may identify promising interventional strategies for improving outcomes in TBI patients clinically.

Abbreviations
- APC
antigen presenting cell
- BBB
blood brain barrier
- CNS
central nervous system
- DAMP
disease‐associated molecular pattern
- IL
interleukin
- MHC
major histocompatibility complex
- MoA
mechanism of action
- S1PR
sphingosine 1‐phosphate receptor
- TBI
traumatic brain injury
- TCR
T cell receptor
- TLR
toll‐like receptor
- Treg
regulatory T cell
- γδ
gamma delta
1. Introduction
Traumatic brain injury (TBI) remains a leading cause of morbidity and mortality across all age groups worldwide [1] and epidemiological studies suggest that TBI prevalence will continue to rise as a result of a growing and aging population [2]. This poses a significant challenge for both afflicted patients, who are burdened with potentially life‐long deficits and an elevated risk of developing comorbid neurological sequelae later in life [3, 4, 5], and society as a whole, which is saddled with the substantial direct and indirect costs associated with TBI management [6]. Nevertheless, no targeted neuroprotective therapies exist to date for mitigating the grave consequences of TBI once head injury has been sustained.
Pathophysiologically, primary mechanical head injury triggers a multifaceted series of cellular and physiological changes that rapidly result in neuroinflammation [7]. Amongst these, central nervous system (CNS) antigens, danger‐associated molecular patterns (DAMPs), and cytokines released from damaged tissue quickly initiate inflammasome activation, which prompts activation and recruitment of CNS resident and peripheral immune cells [8]. Pro‐inflammatory microglial activation and neutrophil infiltration of the CNS occur within hours of primary injury, and peripheral monocytes, natural killer cells, and T cells respond shortly thereafter and continue to exacerbate neuroinflammation [8]. While necessary for clearing damaged tissue and initiating reparative processes, this inflammation also induces secondary brain injury, wherein otherwise viable tissue is harmed [9]. Thus, the detrimental components of neuroinflammation that develop following TBI serve as potential therapeutic targets for intervention.
Unlike many other brain‐infiltrating peripheral immune cells involved in the pathophysiological response to TBI, T cells are known to remain in the brain parenchyma for weeks to months after primary injury [10, 11, 12]. Importantly, T cells are functionally diverse and dynamic, serving both pro‐ and anti‐inflammatory roles in immunological diseases [13]. Evidence from other acute brain injuries has strongly implicated several major T cell populations in aggravating neuroinflammation and contributing to worsened outcomes, including CD8+ [14] and CD4+ [15] T cells. Likewise, in TBI, early evidence has demonstrated that both CD8+ and CD4+ T cells aggravate different aspects of the neuroinflammatory response including microglial activation, blood brain barrier (BBB) permeability, and neuron loss [11, 14, 16], though some subsets including regulatory T cells (Tregs) can have potent anti‐inflammatory functions [17]. Beyond this, however, many critical aspects of the T cell response are poorly understood, and it remains largely unstudied if targeted, T cell‐specific modulation is a viable clinical strategy for reducing secondary injury in TBI.
In this review, we comprehensively discuss the current state of evidence surrounding the role of T cells in TBI. We emphasize the temporospatial characteristics of this response, the mechanisms of T cell activation and recruitment to the CNS post‐TBI, and the ways in which unique T cell subsets exert their immunological functions. We additionally discuss biological and pathological factors that may influence aspects of this process and summarize previous therapeutic strategies that have been shown to alter T cell activity in this setting. We use this framework to identify key gaps in the current literature regarding the role T cells play in the pathophysiology of TBI and to propose clear research priorities for future investigations. These recommendations aim to advance our understanding of this unique adaptive cell population post‐trauma and support the development of effective therapeutic strategies for treating head injuries in the clinic.
2. The T Cell Response to TBI
2.1. Mechanisms of T Cell Activation
T cells are a unique population of adaptive immune cells that are derived from lymphoid progenitor cells and mature into specific functional subsets in the thymus during development before migrating to peripheral storage compartments [18]. In these locations, naïve T cells (e.g., mature T cells that have not previously become activated) remain functionally dormant and surveil the microenvironment in anticipation of immunological cues that induce their activation through several mechanisms. Classically, this activation has been shown to depend on direct interaction between the T cell receptor (TCR) complex on the T cell surface and cognate peptide antigens loaded in either major histocompatibility complex (MHC)‐I or MHC‐II of other cells [19, 20]. Alongside the TCR complex, the co‐receptor CD8 accommodates interaction with MHC‐I, which is expressed on all nucleated cells in mammals, whereas CD4+ T cells interact with MHC‐II, which is exclusively expressed on antigen presenting cells (APCs) [19, 20, 21]. These distinct T cell lineages respond to separate immunogenic cues, with CD8+ T cells primarily responding directly to cells presenting abnormal proteins of endogenous intracellular origin and CD4+ T cells responding to exogenous peptides that have been scavenged from the extracellular compartment and subsequently presented by APCs [20, 21]. However, after sufficient TCR stimulation and co‐stimulation have been achieved in both subtypes (Figure 1), the TCR complex initiates several intracellular signaling cascades that further activate distal transcription factors and other pathways that ultimately direct T cell expansion and phenotypic differentiation depending on the microenvironment's current cytokine signature [22, 23]. This adaptive process is unique in that, unlike in innate immune cell activation, it is highly specific to individual antigenic peptide sequences in a clonal fashion. This pathway, which has recently been shown to play a critical role in potentiating neuroinflammation following intracerebral hemorrhage [24, 25], may be particularly relevant in TBI. In this setting, primary injury instantly mechanically damages neurons and other cells within the CNS when the brain impacts the cranium and rotational forces from the impact are exerted within the parenchyma [26]. This leads to the immediate release of large quantities of potentially immunogenic proteins [27, 28], which, in turn, may amplify the magnitude of TCR‐dependent signaling compared to what is seen following cerebral ischemia, hemorrhage, or other isolated neuroinflammatory diseases. Hence, additional investigations into the role of TCR signaling and the therapeutic potential of its modulation in TBI are warranted.
FIGURE 1.

TCR‐dependent versus independent mechanisms of T cell activation. T cells are classically activated via TCR‐dependent mechanisms (left). Protein antigen is processed by MHC‐expressing antigen presenting cells and subsequently presented extracellularly on either MHC‐I or MHC‐II. The interaction of antigen‐loaded MHC‐I or II with the TCR‐CD3 complex on the surface of T cells and the co‐stimulatory signals CD8 or CD4, respectively, induces intracellular signaling cascades that ultimately promote cellular activation. T cells may also be activated in a TCR‐independent manner (right). Ligand cytokines and DAMPs released by other cells into the intracellular space bind to cytokine receptors or TLRs, respectively. This induces intracellular signaling to promote T cell activation distinct from the pathway initiated by the TCR complex. DAMPs, damage‐associated molecular patterns; MHC, major histocompatibility complex; TCR, T cell receptor; TLR, toll‐like receptor.
In addition to TCR‐dependent activation, recent advances have highlighted the phenomenon of T cell activation occurring independent of TCR complex stimulation. DAMPs, endogenous stress signals released from dead and dying cells, enter the extracellular space and bind with several classes of toll‐like receptors (TLRs) on both innate and adaptive immune cells [29, 30]. DAMPs consist of cellular and extracellular components that are not exposed to surveilling immune cells under homeostatic conditions, including self‐DNA/RNA and structural proteins and peptides [29]. Additionally, cytokines released from other immune cells can directly stimulate cytokine receptors on T cells in a similar manner [31]. Upon successful interaction between DAMP/TLR or cytokine/receptor (Figure 1), numerous intracellular signaling pathways are activated that then promote upregulated transcription of numerous genes, including cytokines and interferons that cause both self‐activation and facilitate activation of neighboring immune cells [29]. As a conserved immunogenic pathway, this stimulation occurs in the absence of T cell interaction with a unique cognate antigen, as is seen through classical TCR‐dependent signaling.
2.2. T Cell Recruitment to the Injury Site
T cell recruitment to the injury site is complex and is an area warranting further scrutiny. It is known that brain resident T cells occupy a vital immunologic role in surveilling for and rapidly responding to brain injury [32, 33]. Nevertheless, it is becoming increasingly clear that peripheral T cell stores also play a major role in this response and are actively recruited into the CNS after acute injury [34, 35]. It has been shown that cytokines and DAMPs are rapidly released from damaged neurons and glia after acute brain injury, and these can interact with immune cells locally but may also travel to the periphery via the meningeal lymphatic system and bloodstream [36, 37, 38]. Once there, DAMPs and cytokines are likely capable of directly stimulating surveilling T cells within the circulation to induce their recruitment to the injury site via TCR‐independent mechanisms. However, because detectable quantities of numerous CNS‐derived antigens are also released into the blood and CSF after TBI [27, 28], it can be hypothesized that naïve T cells in peripheral lymphoid organs like the cervical lymph nodes and spleen may encounter immunogenic CNS antigen after it has been trafficked to the periphery and presented by APCs and subsequently undergo TCR‐dependent activation (Figure 2). Thereafter, activation of these naïve T cells would cause their differentiation, clonal expansion, and egress from the lymphoid tissues into the bloodstream, where they would then home towards the CNS via chemotactic signaling and ultimately infiltrate the parenchyma via the choroid plexus or neurovascular unit [39, 40, 41, 42]. Importantly, it has been shown that disrupting individual steps of this signaling pathway can directly interfere with effective recruitment of T cells to the brain after TBI. For instance, transgenic mice lacking meningeal lymphatics exhibit reduced CD4+ T cell brain infiltration post‐TBI and also lack T cell expansion in the cervical lymph nodes [43]. Cervical lymphadenectomy and cervical afferent lymphatic ligation can also both reduce post‐traumatic CD4+ T cell brain infiltration, reduce cerebral pro‐inflammatory cytokine expression, and improve neurobehavioral performance in mice, amongst other beneficial effects [38]. These suggest that a functionally intact brain to secondary lymphoid organ interface is necessary to effectively coordinate peripheral recruitment of T cells to the injury site, and this may indicate interventions targeting specific steps of this pathway have potential utility for altering the response of key T cell subset populations [38, 40, 43, 44]. Gamma‐delta (γδ) T cells likely respond from atypical lymphoid stores such as the calvarium and meninges in a similar manner [45], but the detailed mechanisms of their response in acute brain injury remain largely unstudied. Additional works are needed to elucidate the respective contributions of peripheral T cell stores versus brain and skull resident T cells to the overall T cell response to TBI. Furthermore, given the existence of multiple, distinct mechanisms of activation, additional investigations that evaluate the subset‐specific involvement of TCR‐dependent versus independent mechanisms focally at sites of neuroinflammation and in the peripheral compartments will be beneficial in TBI, as they may identify novel targets for intervention.
FIGURE 2.

Potential mechanism of TCR‐dependent peripheral T cell recruitment after TBI. After primary mechanical injury, CNS antigens released from the parenchyma enter the meningeal lymphatics and bloodstream (1) where they are trafficked to secondary lymphoid organs like the CLNs and spleen (2). In these sites, APCs process and present the CNS antigen on MHC complexes to directly stimulate naïve T cells via the TCR (3). Chemokines concurrently released from the site of brain injury induce activated T cells to migrate into the bloodstream (4), where they home along the increasing chemokine gradient toward the brain (5) and ultimately infiltrate the parenchyma (6) to reach the site of injury. APCs, antigen presenting cells; CNS, central nervous system; CLNs, cervical lymph nodes; MHC, major histocompatibility complex; TCR, T cell receptor.
2.3. Temporospatial Characteristics of the T Cell Response to TBI
The recovery period following primary injury during TBI can be separated into acute, subacute, and chronic phases. These loosely correspond to the timeframes spanning the first 2–3 days, then out to around 1 month, and then beyond 1 month post‐injury, respectively; though it is important to note that this nomenclature varies across the preclinical and clinical literature. The temporospatial characteristics of T cell activation, differentiation, and recruitment to the CNS after TBI have been most extensively studied using rodent models, where injury parameters and genetic background can be highly controlled (Table 1). However, common trends in the temporospatial characteristics of the T cell response can be observed from the current literature, and studies have demonstrated that the response is dynamic and begins rapidly after TBI induction. In young mice, in which thymic involution has yet to occur, the thymic medullary area has been shown to increase significantly within the acute and early subacute phases [46]. In adult mice, T cells appear to undergo several rounds of proliferation in peripheral lymphoid tissues prior to mobilization during this timeframe as well [40]. Splenic stores of mature T cells then transiently decrease across the subacute phase following primary injury, which likely reflects their mobilization into the bloodstream and gradually decreased expansion [47]. Accordingly, levels of CD4+ and CD8+ T cells in the bloodstream may rise starkly within the acute and early subacute phases post‐injury and gradually return to pre‐injury levels [46, 49]; though this is not always the case, and levels of circulating T cells may remain relatively constant into the subacute phase [48]. One notable caveat lies in that, in severe TBI, peripheral immunosuppression and reduced T cell mobilization may actually be observed in the days that follow primary injury, which may be owed to the inhibition of T cell egress from secondary lymphoid tissues by elevated systemic catecholamine and cortisol levels [56, 57]. Within the brain parenchyma, elevations in the number of brain‐infiltrating T cells can be detected less than an hour after injury [51] and overall T cell infiltration peaks within the first 4 days of the subacute phase [11, 12, 47, 50, 51, 52, 53, 54, 55]. Thereafter, levels of both CD4+ and CD8+ T cells within the brain decrease substantially by around 1 week post‐injury; though several long‐term studies have indicated that levels of both subtypes remain elevated or even begin to rise bimodally well into the chronic phase after injury [10, 11, 12, 47, 53, 54]. Findings from these model studies agree with the limited published literature studying the temporal properties of the T cell response in human subjects [58, 59, 60, 61]. However, several critical components remain undescribed, including what the temporospatial characteristics of specific T cell subsets, including the CD4+ T cell subsets (e.g., Th1, Th2, Th17, Treg) and γδ T cells, are across this time frame and what role injury severity plays in directing the overall polarization of this process. The temporospatial patterns of T cell infiltration within the brain itself after TBI (e.g., T cell infiltration locally at the injury site versus other remote regions of the brain as secondary inflammation progresses) have not been adequately assessed, which may reveal associations between pathological features of TBI and functional outcomes, amongst other significant findings. The dichotomies between continued T cell infiltration versus intraparenchymal expansion and eventual egress versus apoptosis also warrant further scrutiny as they may alter the efficacy of targeting strategies at different time points post‐injury based on the intervention's mechanism of action (MoA).
TABLE 1.
Summary of reported temporospatial observations of the T cell response to TBI in preclinical models.
| Location | Subset | Subjects | Age | Sex | Model | Method | Observation | Ref. |
|---|---|---|---|---|---|---|---|---|
| Thymus | NS | Long Evans Rats | Juvenile (3 wk) | M | CHIMERA | IHC | The thymic medullary area expanded within 2 dpi | [46] |
| Spleen | NS | Long Evans Rats | Juvenile (3 wk) | M | CHIMERA | IHC | There was no difference in germinal center area or primary nodule area at 2 dpi | [46] |
| CD3+ | C57BL/6 Mice | Adult (8–10 wk) | M | CCI | FC | T cell numbers in the spleen fluctuated significantly over the first 7 dpi, remained decreased from 14 to 21 dpi, and returned to baseline at 28 dpi | [47] | |
| CLNs | CD3+ | C57BL/6 Mice | Adult (8–10 wk) | M | CCI | FC | There were significantly more T cells at 28 dpi compared to 1 dpi | [47] |
| PB | CD3+ | Sprague Dawley Rats | Adult (12 wk) | M | CCI–R | FC | Total T cells decreased at 1 dpi, then continually increased to 14 dpi, then decreased below baseline at 28 and 42 dpi | [12] |
| CD8+ | Long Evans Rats | Juvenile (3 wk) | M | CHIMERA | FC | CD8+ T cells were only reduced at 2 h post‐TBI | [46] | |
| Sprague Dawley Rats | Adult (12 wk) | M | CCI–R | FC | Percentage of CD8+ T cells continuously increased from 7 to 42 dpi | [12] | ||
| Sprague Dawley Rats | Adult (NS) | M | bTBI | FC | CD8+ T cells were only reduced at 2 h post‐TBI | [48] | ||
| CD4+ | Long Evans Rats | Juvenile (3 wk) | M | CHIMERA | FC | CD4+ T cells are elevated in the PB at 2 dpi | [46] | |
| Sprague Dawley Rats | Adult (12 wk) | M | CCI–R | FC | Percentage of CD4+ T cells continuously declined from 7 to 42 dpi | [12] | ||
| Sprague Dawley Rats | Adult (NS) | M | bTBI | FC | CD4+ T cells were unchanged out to 3 dpi but reduced at 7 dpi | [48] | ||
| Th2 | C57BL/6 Mice | Adult (NS) | M | CCI | FC | Th2 polarization was unchanged at 1 and 21 dpi but decreased at 3 dpi | [49] | |
| Th17 | C57BL/6 Mice | Adult (NS) | M | CCI | FC | Th17 polarization peaked at 1 dpi and remained elevated at 3 and 21 dpi | [49] | |
| PB | Treg | C57BL/6 Mice | Adult (NS) | M | CCI | FC | Treg numbers were significantly reduced at 1, 3 and 21 dpi | [49] |
| Sprague Dawley Rats | Adult (NS) | M | bTBI | FC | Tregs were only reduced at 3 dpi | [48] | ||
| Brain | CD3+ | Long Evans Rats | Juvenile (3 wk) | M | CHIMERA | FC | There was no difference in total T cells at 2 dpi | [46] |
| C57BL/6 Mice | Adult (8–10 wk) | M | CCI | FC | T cell numbers were transiently elevated across the first 7 dpi and again at 28 dpi, but were not elevated at 14 dpi | [47] | ||
| C57BL/6 Mice | Adult (2 mo) | M | CCI | IHC | T cell numbers were elevated in both the ipsi‐ and contralateral hemispheres at 3 dpi | [50] | ||
| Sprague Dawley Rats | Adult (12 wk) | M | CCI–R | FC | T cells were elevated at 1, 3, 7, 14, 28, and 42 dpi, with distinct peaks at 7 and 42 dpi | [12] | ||
| C57BL/6 Mice | Adult (21 mo) | M | CCI | IHC | T cell numbers were elevated at 3 dpi in the ipsilateral but not the contralateral hemisphere | [50] | ||
| Winstar Rats | Adult (NS) | M | WD–C | IHC | T cell numbers are transiently increased from 5 min to 1 dpi in numerous brain regions | [51] | ||
| C57BL/6 Mice | Adult (NS) | M | cFPI | IHC | T cells were elevated in the perilesional cortex and subcortical white matter at 1 and 3 dpi but not at 7 dpi | [52] | ||
| CD8+ | C57BL/6 Mice | Juvenile (3 wk) | M | CCI | IHC | CD8+ T cells were elevated at 3 and 14 dpi but not 1, 7, or 35 dpi | [53] | |
| C57BL/6 Mice | Adult (9 wk) | M | CCI | qPCR | CD8+ T cells in the cortex peaked at 4 and 14 dpi but remained elevated above baseline from 7 to 35 dpi | [54] | ||
| Sprague Dawley Rats | Adult (12 wk) | M | CCI–R | FC | Percentage of CD8+ T cells decreased to a low at 7 dpi before returning to baseline by 42 dpi | [12] | ||
| C57BL/6 Mice | Adult (12 wk) | M | CCI | IHC | CD8+ T cells were elevated at 3 and 14 dpi but not 1, 7, or 35 dpi | [53] | ||
| C57BL/6 Mice | Adult (7–13 wk) | M | CCI | FC | CD8+ T cells are elevated at 56 dpi | [10] | ||
| Sprague Dawley Rats | NS | M | WD | FC | CD8+ T cell infiltration peaked at 1 dpi but remained elevated through 3 dpi | [55] | ||
| Brain | CD4+ | C57BL/6 Mice | Juvenile (3 wk) | M | CCI | IHC | CD4+ T cells peaked at 1 dpi, remained elevated at 3, 7, and 14 dpi, and returned to baseline at 35 dpi | [53] |
| Sprague Dawley Rats | Adult (12 wk) | M | CCI–R | FC | Percentage of CD4+ T cells rose to a peak at 7 dpi before returning to baseline by 42 dpi | [12] | ||
| C57BL/6 Mice | Adult (12 wk) | M | CCI | IHC | CD4+ T cells were elevated at 1 and 3 dpi but returned to baseline by 7 dpi | [53] | ||
| C57BL/6 Mice | Adult (7–13 wk) | M | CCI | FC | CD4+ T cells were not elevated at 56 dpi | [10] | ||
| Th1 | C57BL/6 Mice | Adult (NS) | M | CCI | FC | Th1 polarization peaked at 1 dpi and were also elevated at 3 and 21 dpi | [49] | |
| Th2 | C57BL/6 Mice | Adult (NS) | M | CCI | FC | Th2 polarization was unchanged at 1 and 7 dpi but was decreased at 3 and 21 dpi | [49] | |
| Th17 | C57BL/6 Mice | Adult (7–13 wk) | M | CCI | FC | Th17 cells were elevated at 7 dpi | [10] | |
| C57BL/6 Mice | Adult (NS) | M | CCI | FC | Th17 polarization peaked at 1 dpi and remained elevated at 3 and 21 dpi | [49] | ||
| Treg | C57BL/6 Mice | Adult (NS) | M | CCI | FC | Treg numbers were significantly reduced at 1, 3 and 21 dpi | [49] |
Abbreviations: bTBI, blast‐induced closed‐head traumatic brain injury; –C, closed‐skull; CCI, controlled cortical impact; cFPI, central fluid percussive injury; CHIMERA, closed head injury model of engineered rotational acceleration; CLNs, cervical lymph nodes; FC, flow cytometry; h, hours; M, male; min, minutes; mo, months; NS, not specified; PB, peripheral blood; qPCR, quantitative polymerase chain reaction; −R, repetitive; WD, weight drop; wk, weeks.
3. Pathological Roles of T Cells in Secondary Brain Injury After TBI
Numerous mechanistic studies have sought to isolate and define the contributions of T cells to the overall post‐traumatic inflammatory milieu from those made by other innate and adaptive cell types. Observational studies have shown that the mere presence of CD3+ T cells, the broadest marker expressed on all T cell populations, within the brain parenchyma post‐TBI is associated with increased microgliosis [11] and corresponds closely with perilesional BBB dysfunction [11, 62]. However, the contributions made by this adaptive immune cell family are complex, given the diverse and dynamic functions of the many T cell subsets (Figure 3) [20]. Hence, there is a need to more finely consider the contributions made by each subset when considering the role and therapeutic potential in TBI.
FIGURE 3.

Contribution of T cell subsets to neuroinflammation in TBI. Upon successful activation, differentiation of naïve CD4+ T cells is directed by the chemokine profile of the microenvironment. Th1 and Th17 helper T cells have potent pro‐inflammatory functions in TBI, whereas Tregs have been shown to attenuate neuroinflammation. The role of Th2 helper T cells remains largely unstudied in TBI to date. CD8+ T cells exert pro‐inflammatory effects, and the functionality of γδ T cells depends on the variable portion of the γ chain of the T cell receptor.
3.1. CD4+ T Cells
Arguably the most phenotypically diverse of the T cell subsets, CD4+ T cells have been shown to make significant contributions to the neuroinflammatory profile that develops after TBI. Effector CD4+ T cells appear to directly aggravate secondary injury in the acute phase, likely due to augmented induction of neuronal apoptosis [38, 63]. As a whole, this suggests CD4+ T cells have a net pro‐inflammatory function in the acute phase; however, upon successful activation of a naïve CD4+ T cell in the presence of the proper microenvironmental cytokine signature, phenotypic differentiation is directed into several distinct subsets including Th1, Th2, Th17, and Treg, each with its own characteristics and functions [23]. Therefore, different CD4+ T cell subsets are likely to have diverse functions after TBI, with some directing pro‐inflammatory polarization and others attenuating this response. For instance, brain infiltrating Th1 cells have been shown to promote sustained microglial pro‐inflammatory activation in the acute phase of TBI via the secretion of IFNγ and TNFα, which is associated with increased demyelination and tissue loss compared to mice subjected to CXCL10 knockdown, a critical chemoattractant for this T cell population [44]. Th17 cells have similarly been shown to contribute to neuroinflammation in TBI, where levels of their effector cytokine interleukin (IL)‐17a are elevated within the first 6 h post‐injury and are associated with increased neuronal apoptosis and overall secondary injury severity [64]; though, surprisingly, one study found that genetic knockout of IL‐17 was not intrinsically neuroprotective [65]. In contrast, Tregs exert potent anti‐inflammatory effects in TBI through the production of IL‐10 and TGFβ, which enable them to directly attenuate the pro‐inflammatory functions of other cells including microglia and other T cells [66]. This function is augmented via the alarmin IL‐33, which has recently been highlighted as an early immune signal released from damaged neurons and glia in acute brain injury [66, 67]. Depletion of Tregs in TBI has detrimental consequences including enhanced brain infiltration of effector T cells, increased reactive astrogliosis, and greater BBB permeability, which contribute to worsened motor performance in the acute phase [68]. Conversely, transfer of umbilical cord blood‐derived Tregs in TBI models has been shown to dramatically reduce neuroinflammation by regulating numerous immune cell populations [69, 70, 71, 72]. Clinically, circulating Treg levels are positively correlated with improved outcomes in TBI patients [73] and severe TBI is associated with impaired Treg function, potentially exacerbating the response of other T cell populations [74]. Taken together, these findings show that the phenotypic differentiation of naïve CD4+ T cells directly influences the pro‐ versus anti‐inflammatory functions of the T cell response to acute brain injury and suggests strategies that aim to reduce effector CD4+ T cell differentiation while promoting Treg expansion may prove beneficial in TBI by augmenting the neuroprotective aspects of this response. Furthermore, while Th2 cells have been shown to be beneficial in other models of acute brain injury [75], to the best of our knowledge, no studies looking into Th2 cells in TBI exist to date, and this is an area in need of additional investigation in the future.
3.2. CD8+ T Cells
To date, there are a limited number of studies that have focused on the function of CD8+ T cells in the recovery period following TBI. While cross‐presentation of exogenous antigen on MHC‐I by APCs to accommodate recognition by CD8+ T cells is a known phenomenon [76], these cells primarily respond to abnormal proteins of intracellular origin, as seen in viral infection [21]. Hence, it remains unclear which immunogenic cues initiate their response in TBI and what mechanisms are responsible for their activation. Functionally, it is known that binding of pro‐inflammatory signals including IL‐15 released from activated astrocytes in response to primary injury can induce CD8+ T cells to secrete granzyme B into the extracellular space [10, 77]. This in turn activates caspase‐3 mediated apoptotic signaling cascades in other local cells including neurons [77]. Notably, this process in TBI is long lasting, as significantly increased levels of CD8+ T cells are detectable in the ipsilateral hemisphere as long as 8 weeks post‐injury in CCI mice [10]. Depletion or genetic deficiency of CD8+ T cells appears to be neuroprotective and is associated with improved neurobehavioral performance, reduced effector CD4+ T cell differentiation and brain infiltration, and reduced white matter injury at chronic timepoints suggesting CD8+ T cells may be a viable target for reducing chronic neuroinflammation after TBI [10]. However, while it has been shown that CD8+ T cells can undergo continued intraparenchymal proliferation after their initial infiltration and persist within the brain for extended periods [40], findings from a recent study suggest that their chronic presence is largely due to continued replenishment from the periphery [78]. This discrepancy is highly translationally relevant as it may predilect certain intervention strategies over others (e.g., targeting peripheral CD8+ T cell activity versus the activity of CD8+ T cells that are already present in the brain) for targeting the chronic neuroinflammation driven by this cell population. In light of this, additional studies that thoroughly investigate both the mechanisms and role of CD8+ T cells and their potential as a therapeutic target after TBI are warranted.
3.3. γδ T Cells
Possessing TCRs comprised of a γ and δ chain (as opposed to that of CD4+ and CD8+ T cells which are comprised of an α and β chain), γδ T cells are non‐conventional in that they are lowly abundant in the peripheral blood and instead are primarily tissue‐resident immune cells that home to peripheral tissues in lieu of traditional secondary lymphoid organs after development in the thymus [79, 80]. γδ T cells express TCRs of relatively low diversity to rapidly respond to distinct cognate antigens, causing them to be described as “innate‐like” and acting as a bridge between innate and antigen‐specific adaptive immune cells [81, 82]. Hence, this T cell population is uniquely poised to act as a first responder to injury within the anatomical compartment in which it resides, including the skull and CNS [79, 83]. However, despite evidence that suggests γδ T cells are important contributors to neuroinflammation in other acute brain injuries [84, 85], little is known about their role in TBI. Indeed, parenchymal γδ T cells have been shown to increase in number within the first 24 h after head injury [86]. Interestingly, TBI was associated with reduced expression of MHC‐II on γδ T cells, which was proposed to be indicative of reduced antigen processing capability and a respective decreased ability to couple innate and adaptive immune responses early after injury, potentially worsening secondary injury [86]. Using γδ knock‐out mice, another study found that γδ T cells worsened neurobehavioral performance in terms of motor function, anxiety‐like behaviors, and memory at chronic time points post‐TBI, though these findings were only significant in male mice [87]. This study also demonstrated that γδ knock‐out mice exhibited reduced acute brain infiltration of both CD4+ and CD8+ T cells, neutrophils, and monocytes, as well as reduced microglial activation at acute and chronic timepoints, supporting the hypothesis that γδ T cells are integral for acutely coordinating both innate and adaptive immune responses after injury. However, the neuroinflammatory functions of γδ T cells were shown to be dependent on the variable portion of the γ chain of the TCR, with Vγ4 γδ T cells potentiating neuroinflammation through the production of both IL‐17 and IFNγ and Vγ1 γδ T cells exerting anti‐inflammatory effects via the expression of TGFβ. Thus, additional works are needed to better understand the functional significance of these paradoxical functions and to further elucidate the dynamic response of this unique T cell subset in TBI.
4. T Cell Interplay With Other Biological and Pathological Factors After TBI
Unlike in models of TBI, which are highly controlled to reproducibly induce homogenous injuries across subjects [88], human TBI is extremely heterogenous [1]. Thus, differences in injury mechanism, location, and severity are known to confound the study of TBI in humans [89] and these factors undoubtedly confound the study of the T cell response clinically. Indeed, preclinical modeling has demonstrated that altering injury mechanism and severity directly influences the cellular and molecular changes that occur following primary injury [90]. However, in addition to primary injury heterogeneity, numerous other factors exist that may also influence the T cell response to TBI.
4.1. Age
This response has recently been shown to be age‐dependent, with the effectiveness of the response decreasing with age [50, 53, 91, 92, 93, 94]. Preclinical evidence has shown that the number of perilesional brain‐infiltrating T cells is higher in young animals compared to aged animals acutely [50], but that T cell infiltration and activation are significantly higher in aged animals at chronic time points [92]. Analysis of age‐dependent changes in the meningeal transcriptome has further shown that aging alone alters the meningeal compartment and primes the adaptive immune system for an exaggerated, pro‐inflammatory response after TBI [91]. These findings corroborate those from several clinical studies, which have shown that younger patients with TBI are more likely to develop autoreactive T cells against myelin basic protein compared to older patients, which is associated with better prognosis and may reflect a protective autoimmune function [94]. Moreover, older patients exhibit altered T cell activation patterns compared to younger patients, with more T cells differentiating into phenotypes with low cytotoxic activity [93]. These may partially explain why older TBI patients have higher morbidity and mortality rates than their younger counterparts [95]. However, further studies are needed to better understand the mechanisms that underlie this association and to determine if age may alter the efficacy of pharmacologically targeting subsets of T cells in TBI recovery, as has been observed in preclinical studies [78, 96].
4.2. Genetics
Host genetics are likely to be highly influential in shaping the T cell response. Allelic differences in the Ciita gene, the master regulator of MHC‐II expression [97], were shown to regulate MHC‐II expression in the injured cortex by controlling mRNA expression of the invariant chain in a rat TBI model [98]. In turn, the degree of MHC‐II expression strongly correlated with the magnitude of T cell infiltration of the brain post‐injury [98]. This is clinically relevant given the highly heterogenous genetic background of human subjects and may explain differences in the T cell response between otherwise similar patients with similar injuries. Studies assessing allelic differences in the Ciita gene and other genes that influence the T cell response in TBI are warranted, and bioinformatics analysis of large omics datasets may be helpful for identifying such targets of interest [99, 100, 101, 102].
4.3. Sexual Dimorphism
Immune cell composition and function vary between biological sexes and are dependent on complex contributions from both sex‐specific hormones and the differential expression of X chromosome‐linked immune‐related genes [103, 104]. In health, it has been shown that basal levels of T cell subsets in the circulation and secondary lymphoid structures differ between male and female subjects [105]. However, this immunological sexual dimorphism also has broad implications across numerous infectious and inflammatory diseases, including acute brain injury [106, 107, 108, 109]. To date, the influence of sex on outcomes after clinical TBI remains controversial; the majority of preclinical studies are modeled in male animals under the justification that TBI is more predominant in male patients [110]. Indeed, in the temporospatial studies reviewed (Table 1), 100% of the data was derived from male animals. Thus, immunological sexual dimorphism in TBI remains largely unexplored, and investigations using both male and female subjects aimed at understanding this phenomenon's potential influence on the T cell response in this setting are likely to provide valuable insights with relevance for therapeutics development.
4.4. Medical Comorbidities
Common metabolic and cardiovascular comorbidities are known to alter T cell homeostasis and function. For example, hypertensive patients show a significant loss of regulatory T cells accompanied by expansion of pro‐inflammatory Th17 cells at baseline [111]. Likewise, type 2 diabetic patients often have chronically elevated Th1 and Th17 effector T cells with concomitant loss of naïve and Treg populations [112]. In dyslipidemia, excess cholesterol can amplify TCR signaling and boost CD8+ T cell effector function, while atherogenic lipids can prime dendritic cells to secrete cytokines that drive differentiation of inflammatory helper T cell subsets [113]. These examples illustrate well‐established links between frequently encountered comorbidities seen in TBI patients and altered T cell function. As a result, these and other comorbidities likely further influence the T cell response to TBI and should be areas of further inquiry.
5. Development of T Cell Targeting Strategies Post–TBI
Numerous studies have investigated the therapeutic potential of pharmacologically targeting secondary injury in TBI, and several of these approaches have been shown to alter the T cell response through both direct and indirect modulation. In general, these approaches aim to either reduce effector T cell functioning or bolster the response of Tregs to reduce neuroinflammation. However, the majority of these studies use treatments with MoAs that are not specific for T cells [44, 55, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125]. For instance, progesterone administration after TBI was recently shown to increase splenic Treg numbers and improve performance on the Morris water maze [114]. Other nonspecific medications including atorvastatin, propofol, dexmedetomidine, doxycycline, and esketamine have also been shown to augment the T cell response to TBI, amongst other effects [115, 116, 121, 122, 125, 126]. Because the MoAs of these medications are not specific to T cells, however, the terminal phenotype of treated subjects undoubtedly receives multifaceted contributions from numerous impacted cell types, and this makes it difficult to isolate potential contributions achieved by altering the T cell response.
Several approaches using interventions with MoAs that target leukocyte activity with higher specificity have been employed in preclinical models of TBI (Table 2). The anti‐α4‐integrin antibody natalizumab, which blocks immune cell diapedesis through the BBB, was recently shown to improve survival and neurobehavioral performance in aged mice at two months post‐injury and, interestingly, augmented the Th2 response in the plasma while reducing the number of CD8+ T cells in the brain at this time point [96]. Further experiments determined that the reduction of CD8+ T cells was a result of their reduced chronic infiltration as opposed to increased resident T cell proliferation, which is consistent with the intervention's peripheral MoA [78]. A similar approach using sphingosine 1‐phosphate receptor (S1PR) antagonists including fingolimod and siponimod, which inhibit lymphocyte egress from lymphoid organs and thereby induce immunosuppression, has also been employed in TBI [130, 131, 132, 133]. These studies found that S1PR antagonism improved a wide array of post‐injury outcome measures including lesion volume, neurobehavioral performance, BBB integrity, cytokine profile, cerebral edema, and neuronal apoptosis, amongst others [130, 131, 133]. This conferred neuroprotection was further shown to occur at least in part through S1PR antagonists' ability to modulate the PI3K/AKT pathway, which regulates several important physiological processes surrounding cell survival [130, 134]. However, an additional study did not identify any neuroprotective benefits of fingolimod after focal cortical cryolesion or weight drop models, which assess for focal and diffuse injury respectively [132]. It is possible that differences in TBI modeling methods confounded these findings, as two of the three studies showing S1PR antagonism was beneficial used the controlled cortical impact model of TBI, which delivers a moderate to severe mechanical injury directly to the exposed cortical surface [131, 133], whereas the focal cortical cryolesion and weight drop models were both performed closed‐skull in the study that found no effect [132]. Thus, differences in injury severity could explain these conflicting findings and studies that investigated this further are likely to be informative.
TABLE 2.
Summary of reported experimental strategies targeting the T cell response in TBI.
| Category | Intervention | Subjects | Observed effects on T cells | Effects on outcomes after TBI | Ref. |
|---|---|---|---|---|---|
| Astrocyte‐secreted IL a | IL‐2 | CCI Mice | ↑ brain Treg percentage | ↓ microglial activation, ↓ lesion volume, ↑ neurobehavioral performance | [127] |
| Calcineurin inhibitor | Cyclosporin A | ACI Mice | Np | ↓ lesion volume, ↓ neuronal apoptosis | [63] |
| Exogenous IL | IL‐2C | CCI Mice | ↑ brain‐infiltrating Tregs, ↑ peripheral/splenic Tregs, ∅ peripheral blood/splenic CD4+ T cells, ∅ peripheral blood/splenic CD8+ T cells | ↓ brain‐infiltrating neutrophils, ↑ neurobehavioral performance, ↑ BBB integrity, ↓ cerebral edema, ↓ pro‐inflammatory cytokines, ↑ anti‐inflammatory cytokines | [128] |
| Exogenous IL | IL‐33 | CCI Mice | ↑ brain‐infiltrating Tregs | ↓ lesion volume, ↑ neurobehavioral performance, ↑ anti‐inflammatory cytokines | [66] |
| MHC‐II Inhibitor | CAP | FPI Mice | ↓ splenic CD4+ T cells, ↓ splenic CD8+ T cells, ↓ splenic Tregs, ↓ γδ T cells | ↓ neurodegeneration, ↓ lesion volume, ↓ pro‐inflammatory cytokines | [86] |
| Monoclonal Antibody | Anti‐α4‐Integrin (Natalizumab) | CCI Mice | ↑ chronic peripheral blood Th2 cells, ↓ chronic brain‐infiltrating CD8+ T cells | ↑ survival, ↑ neurobehavioral performance | [96] |
| Monoclonal Antibody | Anti‐α4‐Integrin (Natalizumab) | CCI Mice | ↓ chronic brain‐infiltrating CD8+ T cells, ↓ total brain T cells | ↑ survival, ↑ neurobehavioral performance | [78] |
| Monoclonal Antibody | Intranasal Anti‐CD3 | CCI Mice | ↑ meninges and brain‐infiltrating Tregs | ↑ neurobehavioral performance, ↓ pro‐inflammatory microglial activation | [129] |
| S1PR Antagonist | Fingolimod (FTY720) | WD Mice | Np | ↑ neurobehavioral performance, ↓ cerebral edema, ↓ lesion volume, ↓ neuronal apoptosis | [130] |
| S1PR Antagonist | Fingolimod (FTY720) | CCI Mice | ↓ brain‐infiltrating T cells, ↑ Treg percentage | ↑ BBB integrity, ↑ neurobehavioral performance, ↓ microglial activation, ↓ brain cytokines | [131] |
| S1PR Antagonist | Fingolimod (FTY720) | FCC Mice & WD Mice | ↓ peripheral blood T cells | ∅ BBB integrity, ∅ neurobehavioral performance, ∅ lesion volume, ∅ neuronal apoptosis | [132] |
| S1PR Antagonist | Siponimod | CCI Mice | ↓ brain‐infiltrating CD4+ T cells, ↓ brain‐infiltrating CD8+ T cells | ↓ microglial activation, ↓ astrocyte activation, ↑ BBB integrity, ↓ lesion volume, ↓ pro‐inflammatory cytokines | [133] |
| S1PR Antagonist | TASP0277308 | CCI Mice | ↓ brain‐infiltrating CD4+ T cells, ↓ brain‐infiltrating CD8+ T cells | ↓ microglial activation, ↓ astrocyte activation, ↑ BBB integrity, ↓ lesion volume, ↓ pro‐inflammatory cytokines | [133] |
Abbreviations: ∅, no difference; ↑, increased; ↓, decreased; ACI, aseptic cerebral injury; BBB, blood brain barrier; CAP, competitive antagonist peptide; CCI, controlled cortical impact; FCC, focal cortical cryolesion; FPI, fluid percussion injury; IL, interleukin; IL‐2C, IL‐2/Anti‐IL‐2 complex; MHC, major histocompatibility complex; Np, not provided; S1PR, sphingosine 1‐phosphate receptor; Treg, regulatory CD4+ T cell; WD, weight drop; γδ, gamma delta.
Via CNS gene delivery platform that causes locally activated astrocytes to secrete IL‐2.
Strategies aimed to specifically target T cells in TBI are less common. The calcineurin inhibitor cyclosporin A has been shown to induce T cell inhibition that is associated with reduced lesion volumes and neuronal apoptosis compared to untreated animals [63]. However, while one clinical study found that cyclosporin A treatment after TBI did not induce further peripheral immunosuppression in severe TBI patients [59], the immunosuppression caused by severe TBI already places these patients at increased risk of severe hospital‐acquired infection [135] and the potential reduction in the body's ability to respond to foreign organisms owed to cyclosporin A treatment limits its practical utility in the clinic. Another approach for T cell inhibition utilized a novel peptide‐based MHC‐II inhibitor that specifically blocks APCs' ability to load processed antigen onto MHC‐II for presentation to surveilling CD4+ T cells, thereby inhibiting their activation [86]. This strategy reduced splenic expansion of several T cell subsets and ultimately reduced neurodegeneration, lesion volumes, and pro‐inflammatory cytokine production within the brain. However, because this approach only mitigates TCR‐dependent activation of CD4+ T cells, it does not address the potentially damaging effects of CD8+ or γδ T cells, which is a limitation of the approach. Alternatively, several groups have focused on exploiting pathways involved in Treg activation and infiltration to augment their anti‐inflammatory properties. For example, treatment with either IL‐33 or IL‐2 has both been shown to be beneficial in TBI as they bolster the function of Tregs to reduce the pro‐inflammatory activation of other immune cells [66, 127, 128]. Translationally, peripheral injection of these cytokines is limited by both their relatively short half‐life in the bloodstream and their potential for causing excess immunosuppression in TBI patients. To overcome this, a novel gene delivery platform that specifically enhances astrocytic expression of IL‐2 only at sites of astrogliosis was developed, which bypasses the periphery and acts primarily on brain‐resident Tregs [127]. When applied in TBI, this was shown to limit microglial activation, reduce lesion volumes, and improve neurobehavioral performance compared to mice receiving gene delivery with the sequence encoding IL‐2 omitted. Another approach utilizing intranasally administered monoclonal anti‐CD3 antibody, which is known to induce differentiation of Tregs [136], was recently shown to increase the number of Tregs that migrate into the injured brain after TBI [129]. IL‐10 secreted from these cells decreased pro‐inflammatory microglial activation and enhanced microglial phagocytic capacity, leading to improved cognitive and motor recovery compared to untreated mice. Collectively, these studies highlight the therapeutic utility of targeting specific components of the T cell response to TBI and warrant further exploration.
6. Limitations of the Current Literature
In addition to the previously discussed limitations inherent in preclinical TBI research introduced by differences in preclinical model selection and cohort selection, several additional important factors should be considered when critically appraising the current literature. It is well established that poor experimental rigor and subsequent reproducibility hampers translational neuroscience research [137]. While certainly not universal (e.g., [87, 127, 129]), many studies reviewed here and within the translational sciences as a whole under‐describe or omit key scientific details that limit the reproducibility of their experimental results and the generalizability of their findings. For instance, the majority of studies included in this review did not provide scientific rationale for sample size selection as a component of the study's methods. Failing to provide such information increases concern for selective interference in data reporting and is avoidable with transparent reporting practices [138]. Moving forward, we emphasize that it is critical for authors to include descriptions of key methodological and interpretation components of their study, including descriptions of statistical analyses performed, rationale for sample size and cohort sex selection, experimenter blinding protocols, subject exclusion criteria, and their own critical appraisal of their study's limitations, amongst others. Doing so will help limit the overinterpretation of future study findings pertaining to the role of T cells in TBI to best facilitate the discovery of viable therapeutics.
7. Future Research Perspectives
Despite the growing body of evidence that has implicated T cells as key players in mediating the neuroinflammatory profile that contributes to secondary brain injury following TBI and demonstrated that targeting T cells in this setting may be a viable therapeutic strategy for limiting neuroinflammation to improve outcomes, several critical areas of focus remain unexplored.
Evidence surrounding the mechanisms of the T cell response to TBI and the diverse ways they interact with other immune and non‐immune cell populations within the brain and meninges remains relatively superficial. To overcome this, publicly available TBI omics datasets (e.g., [139, 140]) have the potential to be used to study cell–cell interaction between T cell subsets and other critical cell populations in this context. A similar approach was recently used to show that CD8+ T cell infiltration of the brain after subarachnoid hemorrhage was dependent on CXCL12 released from vascular endothelial cells and pericytes near the ictus; blockade of this interaction significantly reduces CD8+ T cell infiltration and BBB dysfunction [141]. Applying such methods in TBI and validating identified interactions using complementary molecular techniques including metabolomics and protein‐based assays, amongst others, may elucidate previously unknown interactions between T cell subsets and other cell types like vascular endothelial cells, glia, and neurons in the recovery period after head injury.
It has also not been well established which specific immunogenic cues are responsible for initiating the T cell response, what factors influence their production and release from the CNS, and what resultant mechanisms underlie T cell activation, differentiation, and expansion following head trauma. This is particularly relevant in the context of developing new or repurposing existing therapeutics for application in TBI. The majority of the interventions reviewed herein physically prevent T cells that have already been activated in the periphery from reaching the brain, which successfully blocks effector T cell functioning at the cost of also blocking anti‐inflammatory subsets from being able to dampen the response of other immune cells at the injury site. In contrast, an ideal therapeutic intervention would be subset specific, aiming to selectively prevent the activation of detrimental T cell subsets while bolstering the tolerogenic response of neuroprotective subsets. Considering factors such as the biochemical affinity of antigen to the TCR and the cumulative dose of the TCR‐MHC interaction are known to be involved in shaping the phenotypic differentiation of naïve T cells [142, 143, 144, 145, 146], pharmacologically modulating these initial steps of T cell recruitment post‐injury may be a feasible approach for achieving this. Hence, a deeper understanding of the mechanisms underlying T cell activation and differentiation in TBI is an important area of focus moving forward and may warrant applying single cell RNA sequencing or proteomic techniques to identify how therapeutic treatment alters T cell activation at the signaling level to exert its beneficial effects.
Finally, given that T cells persist in the parenchyma for weeks to months post‐injury [10, 11, 12, 47, 53, 54], characteristics surrounding the longevity of their response may be critical factors to consider when it pertains to therapy development. It remains unknown why T cells do not undergo apoptosis in the time period remote from primary injury like many other responding peripheral immune cells do. Furthermore, it has not been well studied how T cell subset populations within the cortex and bloodstream evolve post‐injury and how this may alter their role in recovery as the majority of the current literature focuses on the T cell response within the acute and subacute periods after injury. It is likely that the T cell landscape undergoes functional transitions at certain timepoints during recovery as it switches from acute pro‐inflammatory functioning toward supporting reparative processes. For instance, in ischemic stroke, pro‐inflammatory T cell subsets including CD8+ and γδ T cells are highly involved in the early pro‐inflammatory response to injury [85, 147]. However, Tregs steadily increase in numbers from 1 to 5 weeks post‐ischemic stroke [148, 149], which supports oligodendrogenesis and white matter repair by promoting tissue‐reparative gene expression in microglia [148]. Similar processes may indeed occur in TBI, and this could underlie the chronic, bimodal increase in T cell numbers within the brain that has been reported in TBI previously. Nevertheless, unraveling this will first require a more granular view of the dynamics of the different T cell subsets' responses to head trauma, which is particularly necessary for T cell populations including Th2 and γδ T cells that are currently understudied in TBI. Analysis using flow cytometry and other cell counting techniques is a logical starting point for achieving this. Understanding the temporal composition of the response is also essential for more finely developing T cell targeting strategies. The reviewed preclinical modulation studies in TBI primarily target acute T cell functioning after primary injury. Special consideration for the temporal trends of specific T cell subsets in the recovery period may identify other critical periods after injury for targeting the activity of individual subsets to better augment the treatment's efficacy and, conversely, may also identify timepoints where pharmacologically altering T cell activity may be harmful.
In summary, while T cells have emerged as key regulators of the neuroimmune response following TBI, a more nuanced understanding of their activation, differentiation, and interactions with other cell populations remains essential. Advancing our knowledge through high‐resolution omics approaches, molecular validation techniques, and longitudinal studies of T cell dynamics will be critical for informing the development of targeted immunomodulatory therapies. Ultimately, tailoring interventions to selectively enhance or suppress specific T cell subsets at appropriate time points post‐injury holds promise for reducing neuroinflammation‐induced secondary injury and ultimately facilitating recovery in TBI patients.
Author Contributions
Mitchell D. Kilgore: conceptualization, methodology, writing – original draft, writing – review and editing, visualization. Yuwen Xiu: conceptualization, methodology, writing – review and editing. Yinghua Jiang: writing – review and editing. Yingjie Wang: writing – review and editing. Mengxuan Shi: writing – review and editing. Di Zhou: writing – review and editing. Thin Sein: writing – review and editing. Sammy J. Vodovoz: writing – review and editing, visualization. Danni Wang: writing – review and editing. Aaron S. Dumont: writing – review and editing, supervision. Aimee Aysenne: writing – review and editing, supervision. Ning Liu: conceptualization, methodology, writing – review and editing, supervision, funding acquisition. Xiaoying Wang: conceptualization, methodology, writing – review and editing, supervision, funding acquisition.
Conflicts of Interest
Dr. Xiaoying Wang is an Editorial Board member of CNS Neuroscience & Therapeutics and a co‐author of this article. To minimize bias, he was excluded from all editorial decision making related to the acceptance of this article for publication. The remaining authors declare no conflicts of interest.
Acknowledgments
Figures were prepared using the tool suite available at BioRender.com [150, 151, 152, 153].
Kilgore M. D., Xiu Y., Jiang Y., et al., “T Cell Involvement in Neuroinflammation After Traumatic Brain Injury: Implications for Therapeutic Intervention,” CNS Neuroscience & Therapeutics 31, no. 8 (2025): e70580, 10.1111/cns.70580.
Funding: National Institutes of Health (NIH) grants (1RO1 NS126503‐01A1to XW), American Heart Association Career Development Award (23CDA1055341 to NL), pilot funding from the Tulane COBRE for Clinical and Translational Research in Cardiometabolic Diseases (to NL), National Institutes of Health (P20GM109036 to NL), and the Tulane School of Medicine Pilot Fund Award (to NL).
Contributor Information
Ning Liu, Email: nliu3@tulane.edu.
Xiaoying Wang, Email: xwang51@tulane.edu.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
References
- 1. Maas A. I. R., Menon D. K., Manley G. T., et al., “Traumatic Brain Injury: Progress and Challenges in Prevention, Clinical Care, and Research,” Lancet Neurology 21, no. 11 (2022): 1004–1060, 10.1016/S1474-4422(22)00309-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Collaborators GTBIaSCI , “Global, Regional, and National Burden of Traumatic Brain Injury and Spinal Cord Injury, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016,” Lancet Neurology 18, no. 1 (2019): 56–87, 10.1016/S1474-4422(18)30415-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Schwarzbold M., Diaz A., Martins E. T., et al., “Psychiatric Disorders and Traumatic Brain Injury,” Neuropsychiatric Disease and Treatment 4, no. 4 (2008): 797–816, 10.2147/ndt.s2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Shively S., Scher A. I., Perl D. P., and Diaz‐Arrastia R., “Dementia Resulting From Traumatic Brain Injury: What Is the Pathology?,” Archives of Neurology 69, no. 10 (2012): 1245–1251, 10.1001/archneurol.2011.3747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Brett B. L., Gardner R. C., Godbout J., Dams‐O'Connor K., and Keene C. D., “Traumatic Brain Injury and Risk of Neurodegenerative Disorder,” Biological Psychiatry 91, no. 5 (2022): 498–507, 10.1016/j.biopsych.2021.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Lo J., Chan L., and Flynn S., “A Systematic Review of the Incidence, Prevalence, Costs, and Activity and Work Limitations of Amputation, Osteoarthritis, Rheumatoid Arthritis, Back Pain, Multiple Sclerosis, Spinal Cord Injury, Stroke, and Traumatic Brain Injury in the United States: A 2019 Update,” Archives of Physical Medicine and Rehabilitation 102, no. 1 (2021): 115–131, 10.1016/j.apmr.2020.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Capizzi A., Woo J., and Verduzco‐Gutierrez M., “Traumatic Brain Injury: An Overview of Epidemiology, Pathophysiology, and Medical Management,” Medical Clinics of North America 104, no. 2 (2020): 213–238, 10.1016/j.mcna.2019.11.001. [DOI] [PubMed] [Google Scholar]
- 8. Needham E. J., Helmy A., Zanier E. R., Jones J. L., Coles A. J., and Menon D. K., “The Immunological Response to Traumatic Brain Injury,” Journal of Neuroimmunology 332 (2019): 112–125, 10.1016/j.jneuroim.2019.04.005. [DOI] [PubMed] [Google Scholar]
- 9. Thapa K., Khan H., Singh T. G., and Kaur A., “Traumatic Brain Injury: Mechanistic Insight on Pathophysiology and Potential Therapeutic Targets,” Journal of Molecular Neuroscience 71, no. 9 (2021): 1725–1742, 10.1007/s12031-021-01841-7. [DOI] [PubMed] [Google Scholar]
- 10. Daglas M., Draxler D. F., Ho H., et al., “Activated CD8+ T Cells Cause Long‐Term Neurological Impairment After Traumatic Brain Injury in Mice,” Cell Reports 29, no. 5 (2019): 1178–1191.e6, 10.1016/j.celrep.2019.09.046. [DOI] [PubMed] [Google Scholar]
- 11. Ndode‐Ekane X. E., Matthiesen L., Bañuelos‐Cabrera I., Palminha C. A. P., and Pitkänen A., “T‐Cell Infiltration Into the Perilesional Cortex Is Long‐Lasting and Associates With Poor Somatomotor Recovery After Experimental Traumatic Brain Injury,” Restorative Neurology and Neuroscience 36, no. 4 (2018): 485–501, 10.3233/RNN-170811. [DOI] [PubMed] [Google Scholar]
- 12. Bai R., Gao H., Han Z., et al., “Flow Cytometric Characterization of T Cell Subsets and Microglia After Repetitive Mild Traumatic Brain Injury in Rats,” Neurochemical Research 42, no. 10 (2017): 2892–2901, 10.1007/s11064-017-2310-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Helsley R. N., Park S. H., Vekaria H. J., et al., “Ketohexokinase‐C Regulates Global Protein Acetylation to Decrease Carnitine Palmitoyltransferase 1a‐Mediated Fatty Acid Oxidation,” Journal of Hepatology 79 (2023): 25–42, 10.1016/j.jhep.2023.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Zhang Z., Duan Z., and Cui Y., “CD8+ T Cells in Brain Injury and Neurodegeneration,” Frontiers in Cellular Neuroscience 17 (2023): 1281763, 10.3389/fncel.2023.1281763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Shi S. X., Xiu Y., Li Y., et al., “CD4+ T Cells Aggravate Hemorrhagic Brain Injury,” Science Advances 9, no. 23 (2023): eabq0712, 10.1126/sciadv.abq0712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Xu L., Ye X., Wang Q., et al., “T‐Cell Infiltration, Contribution and Regulation in the Central Nervous System Post‐Traumatic Injury,” Cell Proliferation 54, no. 8 (2021): e13092, 10.1111/cpr.13092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Shan J., Shi R., Hazra R., and Hu X., “Regulatory T Lymphocytes in Traumatic Brain Injury,” Neurochemistry International 173 (2023): 105660, 10.1016/j.neuint.2023.105660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Kumar B. V., Connors T. J., and Farber D. L., “Human T Cell Development, Localization, and Function Throughout Life,” Immunity 48, no. 2 (2018): 202–213, 10.1016/j.immuni.2018.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Smith‐Garvin J. E., Koretzky G. A., and Jordan M. S., “T Cell Activation,” Annual Review of Immunology 27 (2009): 591–619, 10.1146/annurev.immunol.021908.132706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Sun L., Su Y., Jiao A., Wang X., and Zhang B., “T Cells in Health and Disease,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 235, 10.1038/s41392-023-01471-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Pishesha N., Harmand T. J., and Ploegh H. L., “A Guide to Antigen Processing and Presentation,” Nature Reviews. Immunology 22, no. 12 (2022): 751–764, 10.1038/s41577-022-00707-2. [DOI] [PubMed] [Google Scholar]
- 22. Shah K., Al‐Haidari A., Sun J., et al., “T Cell Receptor (TCR) Signaling in Health and Disease,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 412, 10.1038/s41392-021-00823-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Luckheeram R. V., Zhou R., Verma A. D., and Xia B., “CD4+T Cells: Differentiation and Functions,” Clinical & Developmental Immunology 2012 (2012): 925135, 10.1155/2012/925135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Xiu Y., Wang Y., Wang N., et al., “T Cell Receptor Activation Contributes to Brain Damage After Intracerebral Hemorrhage in Mice,” Journal of Neuroinflammation 22, no. 1 (2025): 78, 10.1186/s12974-025-03402-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Chen S., Fu P., Rastegar‐Kashkooli Y., et al., “AX‐024 Inhibits Antigen‐Specific T‐Cell Response and Improves Intracerebral Hemorrhage Outcomes in Mice,” Stroke 56, no. 5 (2025): 1253–1265, 10.1161/STROKEAHA.124.048507. [DOI] [PubMed] [Google Scholar]
- 26. Dixon K. J., “Pathophysiology of Traumatic Brain Injury,” Physical Medicine and Rehabilitation Clinics of North America 28, no. 2 (2017): 215–225, 10.1016/j.pmr.2016.12.001. [DOI] [PubMed] [Google Scholar]
- 27. Yokobori S., Hosein K., Burks S., Sharma I., Gajavelli S., and Bullock R., “Biomarkers for the Clinical Differential Diagnosis in Traumatic Brain Injury—A Systematic Review,” CNS Neuroscience & Therapeutics 19, no. 8 (2013): 556–565, 10.1111/cns.12127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Ghaith H. S., Nawar A. A., Gabra M. D., et al., “A Literature Review of Traumatic Brain Injury Biomarkers,” Molecular Neurobiology 59, no. 7 (2022): 4141–4158, 10.1007/s12035-022-02822-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Mills K. H., “TLR‐Dependent T Cell Activation in Autoimmunity,” Nature Reviews Immunology 11, no. 12 (2011): 807–822, 10.1038/nri3095. [DOI] [PubMed] [Google Scholar]
- 30. Tang D., Kang R., Coyne C. B., Zeh H. J., and Lotze M. T., “PAMPs and DAMPs: Signal 0s That Spur Autophagy and Immunity,” Immunological Reviews 249, no. 1 (2012): 158–175, 10.1111/j.1600-065X.2012.01146.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Lee H. G., Cho M. J., and Choi J. M., “Bystander CD4+ T Cells: Crossroads Between Innate and Adaptive Immunity,” Experimental & Molecular Medicine 52, no. 8 (2020): 1255–1263, 10.1038/s12276-020-00486-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Liston A., Pasciuto E., Fitzgerald D. C., and Yshii L., “Brain Regulatory T Cells,” Nature Reviews. Immunology 24, no. 5 (2024): 326–337, 10.1038/s41577-023-00960-z. [DOI] [PubMed] [Google Scholar]
- 33. Smolders J., van Luijn M. M., Hsiao C. C., and Hamann J., “T‐Cell Surveillance of the Human Brain in Health and Multiple Sclerosis,” Seminars in Immunopathology 44, no. 6 (2022): 855–867, 10.1007/s00281-022-00926-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Mracsko E., Javidi E., Na S. Y., Kahn A., Liesz A., and Veltkamp R., “Leukocyte Invasion of the Brain After Experimental Intracerebral Hemorrhage in Mice,” Stroke 45, no. 7 (2014): 2107–2114, 10.1161/STROKEAHA.114.005801. [DOI] [PubMed] [Google Scholar]
- 35. Kebir H., Kreymborg K., Ifergan I., et al., “Human TH17 Lymphocytes Promote Blood‐Brain Barrier Disruption and Central Nervous System Inflammation,” Nature Medicine 13, no. 10 (2007): 1173–1175, 10.1038/nm1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Plog B. A., Dashnaw M. L., Hitomi E., et al., “Biomarkers of Traumatic Injury Are Transported From Brain to Blood via the Glymphatic System,” Journal of Neuroscience 35, no. 2 (2015): 518–526, 10.1523/JNEUROSCI.3742-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Javidi E. and Magnus T., “Autoimmunity After Ischemic Stroke and Brain Injury,” Frontiers in Immunology 10 (2019): 686, 10.3389/fimmu.2019.00686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Jiang W., Liu X., Chen Y., et al., “CD4+CD11b+ T Cells Infiltrate and Aggravate the Traumatic Brain Injury Depending on Brain‐to‐Cervical Lymph Node Signaling,” CNS Neuroscience & Therapeutics 30, no. 3 (2024): e14673, 10.1111/cns.14673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Risdall J. E., Carter A. J., Kirkman E., Watts S. A., Taylor C., and Menon D. K., “Endothelial Activation and Chemoattractant Expression Are Early Processes in Isolated Blast Brain Injury,” Neuromolecular Medicine 16, no. 3 (2014): 606–619, 10.1007/s12017-014-8313-y. [DOI] [PubMed] [Google Scholar]
- 40. Ling C., Sandor M., Suresh M., and Fabry Z., “Traumatic Injury and the Presence of Antigen Differentially Contribute to T‐Cell Recruitment in the CNS,” Journal of Neuroscience 26, no. 3 (2006): 731–741, 10.1523/jneurosci.3502-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Marchetti L. and Engelhardt B., “Immune Cell Trafficking Across the Blood‐Brain Barrier in the Absence and Presence of Neuroinflammation,” Vascular Biology 2, no. 1 (2020): H1–H18, 10.1530/VB-19-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Shi S. X., Vodovoz S. J., Xiu Y., et al., “T‐Lymphocyte Interactions With the Neurovascular Unit: Implications in Intracerebral Hemorrhage,” Cells 11, no. 13 (2022): 2011, 10.3390/cells11132011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Wojciechowski S., Virenque A., Vihma M., et al., “Developmental Dysfunction of the Central Nervous System Lymphatics Modulates the Adaptive Neuro‐Immune Response in the Perilesional Cortex in a Mouse Model of Traumatic Brain Injury,” Frontiers in Immunology 11 (2020): 559810, 10.3389/fimmu.2020.559810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Sen T., Saha P., Gupta R., et al., “Aberrant ER Stress Induced Neuronal‐IFNβ Elicits White Matter Injury due to Microglial Activation and T‐Cell Infiltration After TBI,” Journal of Neuroscience 40, no. 2 (2020): 424–446, 10.1523/jneurosci.0718-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Park J. H., Kang I., and Lee H. K., “γδ T Cells in Brain Homeostasis and Diseases,” Frontiers in Immunology 13 (2022): 886397, 10.3389/fimmu.2022.886397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Smith A. M., Taylor E. B., Brooks R. J., dos Santos e Santos C., and Grayson B. E., “Cerebral and Peripheral Immune Cell Changes Following Rodent Juvenile Traumatic Brain Injury,” Brain Sciences 14, no. 4 (2024): 398, 10.3390/brainsci14040398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Jin X., Ishii H., Bai Z., Itokazu T., and 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 7, no. 7 (2012): e41892, 10.1371/journal.pone.0041892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Shi Q. X., Chen B., Nie C., et al., “A Novel Model of Blast Induced Traumatic Brain Injury Caused by Compressed Gas Produced Sustained Cognitive Deficits in Rats: Involvement of Phosphorylation of Tau at the Thr205 Epitope,” Brain Research Bulletin 157 (2020): 149–161, 10.1016/j.brainresbull.2020.02.002. [DOI] [PubMed] [Google Scholar]
- 49. Braun M., Vaibhav K., Saad N., et al., “Activation of Myeloid TLR4 Mediates T Lymphocyte Polarization After Traumatic Brain Injury,” Journal of Immunology 198, no. 9 (2017): 3615–3626, 10.4049/jimmunol.1601948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Timaru‐Kast R., Luh C., Gotthardt P., et al., “Influence of Age on Brain Edema Formation, Secondary Brain Damage and Inflammatory Response After Brain Trauma in Mice,” PLoS One 7, no. 8 (2012): e43829, 10.1371/journal.pone.0043829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Czigner A., Mihály A., Farkas O., et al., “Kinetics of the Cellular Immune Response Following Closed Head Injury,” Acta Neurochirurgica 149, no. 3 (2007): 281–289, 10.1007/s00701-006-1095-8. [DOI] [PubMed] [Google Scholar]
- 52. Ekmark‐Lewén S., Flygt J., Kiwanuka O., et al., “Traumatic Axonal Injury in the Mouse Is Accompanied by a Dynamic Inflammatory Response, Astroglial Reactivity and Complex Behavioral Changes,” Journal of Neuroinflammation 10 (2013): 44, 10.1186/1742-2094-10-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Claus C. P., Tsuru‐Aoyagi K., Adwanikar H., Walker B., Whetstone W., and Noble‐Haeusslein L. J., “Age Is a Determinant of Leukocyte Infiltration and Loss of Cortical Volume After Traumatic Brain Injury,” Developmental Neuroscience 32, no. 5–6 (2010): 454–465, 10.1159/000316805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Ciechanowska A., Ciapała K., Pawlik K., et al., “Initiators of Classical and Lectin Complement Pathways Are Differently Engaged After Traumatic Brain Injury‐Time‐Dependent Changes in the Cortex, Striatum, Thalamus and Hippocampus in a Mouse Model,” International Journal of Molecular Sciences 22, no. 1 (2020): 45, 10.3390/ijms22010045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Clausen F., Lorant T., Lewén A., and Hillered L., “T Lymphocyte Trafficking: A Novel Target for Neuroprotection in Traumatic Brain Injury,” Journal of Neurotrauma 24, no. 8 (2007): 1295–1307, 10.1089/neu.2006.0258. [DOI] [PubMed] [Google Scholar]
- 56. Dong T., Zhi L., Bhayana B., and Wu M. X., “Cortisol‐Induced Immune Suppression by a Blockade of Lymphocyte Egress in Traumatic Brain Injury,” Journal of Neuroinflammation 13, no. 1 (2016): 197, 10.1186/s12974-016-0663-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Yang Y., Ye Y., Chen C., et al., “Acute Traumatic Brain Injury Induces CD4+ and CD8+ T Cell Functional Impairment by Upregulating the Expression of PD‐1 via the Activated Sympathetic Nervous System,” Neuroimmunomodulation 26, no. 1 (2019): 43–57, 10.1159/000495465. [DOI] [PubMed] [Google Scholar]
- 58. Mrakovcic‐Sutic I., Tokmadzic V. S., Laskarin G., et al., “Early Changes in Frequency of Peripheral Blood Lymphocyte Subpopulations in Severe Traumatic Brain‐Injured Patients,” Scandinavian Journal of Immunology 72, no. 1 (2010): 57–65, 10.1111/j.1365-3083.2010.02407.x. [DOI] [PubMed] [Google Scholar]
- 59. Mazzeo A. T., Kunene N. K., Gilman C. B., Hamm R. J., Hafez N., and Bullock M. R., “Severe Human Traumatic Brain Injury, but not Cyclosporin a Treatment, Depresses Activated T Lymphocytes Early After Injury,” Journal of Neurotrauma 23, no. 6 (2006): 962–975, 10.1089/neu.2006.23.962. [DOI] [PubMed] [Google Scholar]
- 60. Ditsch A., Hunold L., Hefele F., et al., “Traumatic Brain Injury Induces a Differential Immune Response in Polytrauma Patients; Prospective Analysis of CD69 Expression on T Cells and Platelet Expansion,” Journal of Clinical Medicine 11, no. 18 (2022): 5315, 10.3390/jcm11185315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Holmin S., Söderlund J., Biberfeld P., et al., “Intracerebral Inflammation After Human Brain Contusion,” Neurosurgery 42, no. 2 (1998): 291–298, 10.1097/00006123-199802000-00047. [DOI] [PubMed] [Google Scholar]
- 62. Holmin S., Mathiesen T., Shetye J., and Biberfeld P., “Intracerebral Inflammatory Response to Experimental Brain Contusion,” Acta Neurochirurgica 132, no. 1–3 (1995): 110–119, 10.1007/BF01404857. [DOI] [PubMed] [Google Scholar]
- 63. Fee D., Crumbaugh A., Jacques T., et al., “Activated/Effector CD4+ T Cells Exacerbate Acute Damage in the Central Nervous System Following Traumatic Injury,” Journal of Neuroimmunology 136, no. 1–2 (2003): 54–66, 10.1016/s0165-5728(03)00008-0. [DOI] [PubMed] [Google Scholar]
- 64. Li T., Zhang Y. M., Han D., et al., “Involvement of IL‐17 in Secondary Brain Injury After a Traumatic Brain Injury in Rats,” Neuromolecular Medicine 19, no. 4 (2017): 541–554, 10.1007/s12017-017-8468-4. [DOI] [PubMed] [Google Scholar]
- 65. Simon D. W., Raphael I., Johnson K. M., et al., “Endogenous Interleukin‐17a Contributes to Normal Spatial Memory Retention but Does Not Affect Early Behavioral or Neuropathological Outcomes After Experimental Traumatic Brain Injury,” Neurotrauma Reports 3, no. 1 (2022): 340–351, 10.1089/neur.2022.0017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Xie D., Miao W., Xu F., et al., “IL‐33/ST2 Axis Protects Against Traumatic Brain Injury Through Enhancing the Function of Regulatory T Cells,” Frontiers in Immunology 13 (2022): 860772, 10.3389/fimmu.2022.860772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Gadani S. P., Walsh J. T., Smirnov I., Zheng J., and Kipnis J., “The Glia‐Derived Alarmin IL‐33 Orchestrates the Immune Response and Promotes Recovery Following CNS Injury,” Neuron 85, no. 4 (2015): 703–709, 10.1016/j.neuron.2015.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Krämer T. J., Hack N., Brühl T. J., et al., “Depletion of Regulatory T Cells Increases T Cell Brain Infiltration, Reactive Astrogliosis, and Interferon‐γ Gene Expression in Acute Experimental Traumatic Brain Injury,” Journal of Neuroinflammation 16, no. 1 (2019): 163, 10.1186/s12974-019-1550-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Caplan H. W., Prabhakara K. S., Kumar A., et al., “Human Cord Blood‐Derived Regulatory T‐Cell Therapy Modulates the Central and Peripheral Immune Response After Traumatic Brain Injury,” Stem Cells Translational Medicine 9, no. 8 (2020): 903–916, 10.1002/sctm.19-0444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Caplan H. W., Prabhakara K. S., Toledano Furman N. E., et al., “Combination Therapy With Treg and Mesenchymal Stromal Cells Enhances Potency and Attenuation of Inflammation After Traumatic Brain Injury Compared to Monotherapy,” Stem Cells 39, no. 3 (2021): 358–370, 10.1002/stem.3320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Caplan H. W., Prabhakara K. S., Toledano Furman N. E., et al., “Human‐Derived Treg and MSC Combination Therapy May Augment Immunosuppressive Potency In Vitro, but Did not Improve Blood Brain Barrier Integrity in an Experimental Rat Traumatic Brain Injury Model,” PLoS One 16, no. 5 (2021): e0251601, 10.1371/journal.pone.0251601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Chen C., Hu N., Wang J., et al., “Umbilical Cord Mesenchymal Stem Cells Promote Neurological Repair After Traumatic Brain Injury Through Regulating Treg/Th17 Balance,” Brain Research 1775 (2022): 147711, 10.1016/j.brainres.2021.147711. [DOI] [PubMed] [Google Scholar]
- 73. Li M., Lin Y. P., Chen J. L., Li H., Jiang R. C., and Zhang J. N., “Role of Regulatory T Cell in Clinical Outcome of Traumatic Brain Injury,” Chinese Medical Journal 128, no. 8 (2015): 1072–1078, 10.4103/0366-6999.155094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Quattrocchi K. B., Issel B. W., Miller C. H., et al., “Impairment of Helper T‐Cell Function Following Severe Head Injury,” Journal of Neurotrauma 9, no. 1 (1992): 1–9, 10.1089/neu.1992.9.1. [DOI] [PubMed] [Google Scholar]
- 75. Wolf S. A., Fisher J., Bechmann I., Steiner B., Kwidzinski E., and Nitsch R., “Neuroprotection by T‐Cells Depends on Their Subtype and Activation State,” Journal of Neuroimmunology 133, no. 1–2 (2002): 72–80, 10.1016/s0165-5728(02)00367-3. [DOI] [PubMed] [Google Scholar]
- 76. Cruz F. M., Colbert J. D., Merino E., Kriegsman B. A., and Rock K. L., “The Biology and Underlying Mechanisms of Cross‐Presentation of Exogenous Antigens on MHC‐I Molecules,” Annual Review of Immunology 35 (2017): 149–176, 10.1146/annurev-immunol-041015-055254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wu L., Ji N. N., Wang H., et al., “Domino Effect of Interleukin‐15 and CD8 T‐Cell‐Mediated Neuronal Apoptosis in Experimental Traumatic Brain Injury,” Journal of Neurotrauma 38, no. 10 (2021): 1450–1463, 10.1089/neu.2017.5607. [DOI] [PubMed] [Google Scholar]
- 78. Chen Z., Ford K. P., Islam M. B. A. R., et al., “Anti‐CD49d Ab Treatment Ameliorates Age‐Associated Inflammatory Response and Mitigates CD8+ T‐Cell Cytotoxicity After Traumatic Brain Injury,” Journal of Neuroinflammation 21, no. 1 (2024): 267, 10.1186/s12974-024-03257-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Vantourout P. and Hayday A., “Six‐of‐the‐Best: Unique Contributions of γδ T Cells to Immunology,” Nature Reviews. Immunology 13, no. 2 (2013): 88–100, 10.1038/nri3384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ribot J. C., Lopes N., and Silva‐Santos B., “γδ T Cells in Tissue Physiology and Surveillance,” Nature Reviews. Immunology 21, no. 4 (2021): 221–232, 10.1038/s41577-020-00452-4. [DOI] [PubMed] [Google Scholar]
- 81. Pellicci D. G., Koay H. F., and Berzins S. P., “Thymic Development of Unconventional T Cells: How NKT Cells, MAIT Cells and γδ T Cells Emerge,” Nature Reviews. Immunology 20, no. 12 (2020): 756–770, 10.1038/s41577-020-0345-y. [DOI] [PubMed] [Google Scholar]
- 82. Lv M., Zhang Z., and Cui Y., “Unconventional T Cells in Brain Homeostasis, Injury and Neurodegeneration,” Frontiers in Immunology 14 (2023): 1273459, 10.3389/fimmu.2023.1273459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Wo J., Zhang F., Li Z., Sun C., Zhang W., and Sun G., “The Role of Gamma‐Delta T Cells in Diseases of the Central Nervous System,” Frontiers in Immunology 11 (2020): 580304, 10.3389/fimmu.2020.580304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Sun G., Yang S., Cao G., et al., “γδ T Cells Provide the Early Source of IFN‐γ to Aggravate Lesions in Spinal Cord Injury,” Journal of Experimental Medicine 215, no. 2 (2018): 521–535, 10.1084/jem.20170686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Shichita T., Sugiyama Y., Ooboshi H., et al., “Pivotal Role of Cerebral Interleukin‐17‐Producing gammadeltaT Cells in the Delayed Phase of Ischemic Brain Injury,” Nature Medicine 15, no. 8 (2009): 946–950, 10.1038/nm.1999. [DOI] [PubMed] [Google Scholar]
- 86. Tobin R. P., Mukherjee S., Kain J. M., et al., “Traumatic Brain Injury Causes Selective, CD74‐Dependent Peripheral Lymphocyte Activation That Exacerbates Neurodegeneration,” Acta Neuropathologica Communications 2 (2014): 143, 10.1186/s40478-014-0143-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Abou‐El‐Hassan H., Rezende R. M., Izzy S., et al., “Vγ1 and Vγ4 Gamma‐Delta T Cells Play Opposing Roles in the Immunopathology of Traumatic Brain Injury in Males,” Nature Communications 14, no. 1 (2023): 4286, 10.1038/s41467-023-39857-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Fesharaki‐Zadeh A. and Datta D., “An Overview of Preclinical Models of Traumatic Brain Injury (TBI): Relevance to Pathophysiological Mechanisms,” Frontiers in Cellular Neuroscience 18 (2024): 1371213, 10.3389/fncel.2024.1371213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Rubiano A. M. and Lee K., “Traumatic Brain Injury Research: Homogenising Heterogeneity,” Lancet Neurology 22, no. 11 (2023): 973–975, 10.1016/S1474-4422(23)00325-3. [DOI] [PubMed] [Google Scholar]
- 90. McDonald B. Z., Tarudji A. W., Zhang H., et al., “Traumatic Brain Injury Heterogeneity Affects Cell Death and Autophagy,” Experimental Brain Research 242, no. 7 (2024): 1645–1658, 10.1007/s00221-024-06856-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Buenaventura R. G., Harvey A. C., Burns M. P., and Main B. S., “Traumatic Brain Injury Induces an Adaptive Immune Response in the Meningeal Transcriptome That Is Amplified by Aging,” Frontiers in Neuroscience 17 (2023): 1210175, 10.3389/fnins.2023.1210175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Chen Z., Islam M., Ford K. P., et al., “Microglia and Infiltrating T‐Cells Adopt Long‐Term, Age‐Specific, Transcriptional Changes After Traumatic Brain Injury in Mice,” Shock 59, no. 2 (2023): 267–276, 10.1097/shk.0000000000002027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Magatti M., Pischiutta F., Ortolano F., et al., “Systemic Immune Response in Young and Elderly Patients After Traumatic Brain Injury,” Immunity & Ageing 20, no. 1 (2023): 41, 10.1186/s12979-023-00369-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Cox A. L., Coles A. J., Nortje J., et al., “An Investigation of Auto‐Reactivity After Head Injury,” Journal of Neuroimmunology 174, no. 1–2 (2006): 180–186, 10.1016/j.jneuroim.2006.01.007. [DOI] [PubMed] [Google Scholar]
- 95. Maiden M. J., Cameron P. A., Rosenfeld J. V., Cooper D. J., McLellan S., and Gabbe B. J., “Long‐Term Outcomes After Severe Traumatic Brain Injury in Older Adults. A Registry‐Based Cohort Study,” American Journal of Respiratory and Critical Care Medicine 201, no. 2 (2020): 167–177, 10.1164/rccm.201903-0673OC. [DOI] [PubMed] [Google Scholar]
- 96. Chen Z., Ford K., Islam M., et al., “Anti‐CD49d Antibody Treatment Improves Survival and Attenuates Neurocognitive Deficits After Traumatic Brain Injury in Aged Mice,” Shock 61 (2023): 112–119, 10.1097/shk.0000000000002256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. León Machado J. A. and Steimle V., “The MHC Class II Transactivator CIITA Not (Quite) the Odd‐One‐Out Anymore Among NLR Proteins,” International Journal of Molecular Sciences 22, no. 3 (2021): 1074, 10.3390/ijms22031074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Al Nimer F., Beyeen A. D., Lindblom R., et al., “Both MHC and Non‐MHC Genes Regulate Inflammation and T‐Cell Response After Traumatic Brain Injury,” Brain, Behavior, and Immunity 25, no. 5 (2011): 981–990, 10.1016/j.bbi.2010.10.017. [DOI] [PubMed] [Google Scholar]
- 99. Yan L., Han X., Zhang M., et al., “Integrative Analysis of TBI Data Reveals Lgmn as a Key Player in Immune Cell‐Mediated Ferroptosis,” BMC Genomics 24, no. 1 (2023): 747, 10.1186/s12864-023-09842-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Zhao G., Fu Y., Yang C., Yang X., and Hu X., “CASP8 Is a Potential Therapeutic Target and Is Correlated With Pyroptosis in Traumatic Brain Injury,” World Neurosurgery 174 (2023): e103–e117, 10.1016/j.wneu.2023.03.004. [DOI] [PubMed] [Google Scholar]
- 101. Qian F., Zhong Q., and Chen Z., “Role of Mitochondrial Dysfunction in Acute Traumatic Brain Injury: Evidence From Bioinformatics Analysis,” Heliyon 10, no. 10 (2024): e31121, 10.1016/j.heliyon.2024.e31121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Ren B., Liang J., Yang L., Wei X., Guo M., and Li H., “Bioinformatics‐Driven Exploration of Key Genes and Mechanisms Underlying Oxidative Stress in Traumatic Brain Injury,” Frontiers in Aging Neuroscience 17 (2025): 1531317, 10.3389/fnagi.2025.1531317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Gal‐Oz S. T. and Shay T., “Immune Sexual Dimorphism: Connecting the Dots,” Physiology 37, no. 2 (2022): 55–68, 10.1152/physiol.00006.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Forsyth K. S., Jiwrajka N., Lovell C. D., Toothacre N. E., and Anguera M. C., “The conneXion Between Sex and Immune Responses,” Nature Reviews. Immunology 24, no. 7 (2024): 487–502, 10.1038/s41577-024-00996-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Dodd K. C. and Menon M., “Sex Bias in Lymphocytes: Implications for Autoimmune Diseases,” Frontiers in Immunology 13 (2022): 945762, 10.3389/fimmu.2022.945762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Liu J., Sato Y., Falcone‐Juengert J., Kurisu K., Shi J., and Yenari M. A., “Sexual Dimorphism in Immune Cell Responses Following Stroke,” Neurobiology of Disease 172 (2022): 105836, 10.1016/j.nbd.2022.105836. [DOI] [PubMed] [Google Scholar]
- 107. Banerjee A. and McCullough L. D., “Sex‐Specific Immune Responses in Stroke,” Stroke 53, no. 5 (2022): 1449–1459, 10.1161/STROKEAHA.122.036945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Kim T., Chelluboina B., Chokkalla A. K., and Vemuganti R., “Age and Sex Differences in the Pathophysiology of Acute CNS Injury,” Neurochemistry International 127 (2019): 22–28, 10.1016/j.neuint.2019.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Bravo‐Alegria J., McCullough L. D., and Liu F., “Sex Differences in Stroke Across the Lifespan: The Role of T Lymphocytes,” Neurochemistry International 107 (2017): 127–137, 10.1016/j.neuint.2017.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Späni C. B., Braun D. J., and Van Eldik L. J., “Sex‐Related Responses After Traumatic Brain Injury: Considerations for Preclinical Modeling,” Frontiers in Neuroendocrinology 50 (2018): 52–66, 10.1016/j.yfrne.2018.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Fu M., Lv M., Guo J., et al., “The Clinical Significance of T‐Cell Regulation in Hypertension Treatment,” Frontiers in Immunology 16 (2025): 1550206, 10.3389/fimmu.2025.1550206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Xia C., Rao X., and Zhong J., “Role of T Lymphocytes in Type 2 Diabetes and Diabetes‐Associated Inflammation,” Journal Diabetes Research 2017 (2017): 6494795, 10.1155/2017/6494795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Kim D., Chung H., Lee J. E., Kim J., Hwang J., and Chung Y., “Immunologic Aspects of Dyslipidemia: A Critical Regulator of Adaptive Immunity and Immune Disorders,” Journal of Lipid and Atherosclerosis 10, no. 2 (2021): 184–201, 10.12997/jla.2021.10.2.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Zhou Z., Li Y., Peng R., et al., “Progesterone Induces Neuroprotection Associated With Immune/Inflammatory Modulation in Experimental Traumatic Brain Injury,” Neuroreport 35, no. 6 (2024): 352–360, 10.1097/WNR.0000000000002013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Xu X., Gao W., Cheng S., et al., “Anti‐Inflammatory and Immunomodulatory Mechanisms of Atorvastatin in a Murine Model of Traumatic Brain Injury,” Journal of Neuroinflammation 14, no. 1 (2017): 167, 10.1186/s12974-017-0934-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Karakaya D., Cakir‐Aktas C., Uzun S., Soylemezoglu F., and Mut M., “Tailored Therapeutic Doses of Dexmedetomidine in Evolving Neuroinflammation After Traumatic Brain Injury,” Neurocritical Care 36, no. 3 (2022): 802–814, 10.1007/s12028-021-01381-3. [DOI] [PubMed] [Google Scholar]
- 117. Farr S. A., Cuzzocrea S., Esposito E., et al., “Adenosine A3 Receptor as a Novel Therapeutic Target to Reduce Secondary Events and Improve Neurocognitive Functions Following Traumatic Brain Injury,” Journal of Neuroinflammation 17, no. 1 (2020): 339, 10.1186/s12974-020-02009-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Appel D., Hummel R., Weidemeier M., Endres K., Gölz C., and Schäfer M. K. E., “Pharmacologic Inhibition of ADAM10 Attenuates Brain Tissue Loss, Axonal Injury and Pro‐Inflammatory Gene Expression Following Traumatic Brain Injury in Mice,” Frontiers in Cell and Development Biology 9 (2021): 661462, 10.3389/fcell.2021.661462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Hua R., Mao S. S., Zhang Y. M., Chen F. X., Zhou Z. H., and Liu J. Q., “Effects of Pituitary Adenylate Cyclase Activating Polypeptide on CD4(+)/CD8(+) T Cell Levels After Traumatic Brain Injury in a Rat Model,” World Journal of Emergency Medicine 3, no. 4 (2012): 294–298, 10.5847/wjem.j.issn.1920-8642.2012.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Clausen F., Hånell A., Björk M., et al., “Neutralization of Interleukin‐1beta Modifies the Inflammatory Response and Improves Histological and Cognitive Outcome Following Traumatic Brain Injury in Mice,” European Journal of Neuroscience 30, no. 3 (2009): 385–396, 10.1111/j.1460-9568.2009.06820.x. [DOI] [PubMed] [Google Scholar]
- 121. Marjani S., Zirh S., Sever‐Bahcekapili M., Cakir‐Aktas C., Muftuoglu S. F., and Mut M., “Doxycycline Alleviates Acute Traumatic Brain Injury by Suppressing Neuroinflammation and Apoptosis in a Mouse Model,” Journal of Neuroimmunology 359 (2021): 577672, 10.1016/j.jneuroim.2021.577672. [DOI] [PubMed] [Google Scholar]
- 122. Cui C., Zhang D., Sun K., et al., “Propofol Maintains Th17/Treg Cell Balance and Reduces Inflammation in Rats With Traumatic Brain Injury via the miR‐145‐3p/NFATc2/NF‐κB Axis,” International Journal of Molecular Medicine 48, no. 1 (2021): 135, 10.3892/ijmm.2021.4968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Shi Q. X., Chen B., Nie C., et al., “Improvement in Cognitive Dysfunction Following Blast Induced Traumatic Brain Injury by Thymosin α1 in Rats: Involvement of Inhibition of Tau Phosphorylation at the Thr205 Epitope,” Brain Research 1747 (2020): 147038, 10.1016/j.brainres.2020.147038. [DOI] [PubMed] [Google Scholar]
- 124. Zhou Z. W., Li F., Zheng Z. T., et al., “Erythropoietin Regulates Immune/Inflammatory Reaction and Improves Neurological Function Outcomes in Traumatic Brain Injury,” Brain and Behavior: A Cognitive Neuroscience Perspective 7, no. 11 (2017): e00827, 10.1002/brb3.827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Liu Y., Gong Z., Zhang L., et al., “Esketamine Attenuates Traumatic Brain Injury by Modulating STAT3‐Mediated Glycolysis and Immune Responses,” BMC Neuroscience 26, no. 1 (2025): 21, 10.1186/s12868-025-00941-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Wang D., Sun H., Hai K., Li N., Gu Y., and Ma Z., “Propofol Alleviates Traumatic Brain Injury Through Regulating Th17/Treg Balance by Activation of the AMPK/SIRT1 Pathway,” Toxicology Mechanisms and Methods 35 (2025): 1–11, 10.1080/15376516.2025.2481893. [DOI] [PubMed] [Google Scholar]
- 127. Yshii L., Pasciuto E., Bielefeld P., et al., “Astrocyte‐Targeted Gene Delivery of Interleukin 2 Specifically Increases Brain‐Resident Regulatory T Cell Numbers and Protects Against Pathological Neuroinflammation,” Nature Immunology 23, no. 6 (2022): 878–891, 10.1038/s41590-022-01208-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Gao W., Li F., Zhou Z., et al., “IL‐2/Anti‐IL‐2 Complex Attenuates Inflammation and BBB Disruption in Mice Subjected to Traumatic Brain Injury,” Frontiers in Neurology 8 (2017): 281, 10.3389/fneur.2017.00281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Izzy S., Yahya T., Albastaki O., et al., “Nasal Anti‐CD3 Monoclonal Antibody Ameliorates Traumatic Brain Injury, Enhances Microglial Phagocytosis and Reduces Neuroinflammation via IL‐10‐Dependent T,” Nature Neuroscience 28, no. 3 (2025): 499–516, 10.1038/s41593-025-01877-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Zhang L., Ding K., Wang H., Wu Y., and Xu J., “Traumatic Brain Injury‐Induced Neuronal Apoptosis Is Reduced Through Modulation of PI3K and Autophagy Pathways in Mouse by FTY720,” Cellular and Molecular Neurobiology 36, no. 1 (2016): 131–142, 10.1007/s10571-015-0227-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Gao C., Qian Y., Huang J., et al., “A Three‐Day Consecutive Fingolimod Administration Improves Neurological Functions and Modulates Multiple Immune Responses of CCI Mice,” Molecular Neurobiology 54, no. 10 (2017): 8348–8360, 10.1007/s12035-016-0318-0. [DOI] [PubMed] [Google Scholar]
- 132. Mencl S., Hennig N., Hopp S., et al., “FTY720 Does not Protect From Traumatic Brain Injury in Mice Despite Reducing Posttraumatic Inflammation,” Journal of Neuroimmunology 274, no. 1–2 (2014): 125–131, 10.1016/j.jneuroim.2014.07.010. [DOI] [PubMed] [Google Scholar]
- 133. Cuzzocrea S., Doyle T., Campolo M., et al., “Sphingosine 1‐Phosphate Receptor Subtype 1 as a Therapeutic Target for Brain Trauma,” Journal of Neurotrauma 35, no. 13 (2018): 1452–1466, 10.1089/neu.2017.5391. [DOI] [PubMed] [Google Scholar]
- 134. Yap T. A., Garrett M. D., Walton M. I., Raynaud F., de Bono J. S., and Workman P., “Targeting the PI3K‐AKT‐mTOR Pathway: Progress, Pitfalls, and Promises,” Current Opinion in Pharmacology 8, no. 4 (2008): 393–412, 10.1016/j.coph.2008.08.004. [DOI] [PubMed] [Google Scholar]
- 135. Sharma R., Shultz S. R., Robinson M. J., et al., “Infections After a Traumatic Brain Injury: The Complex Interplay Between the Immune and Neurological Systems,” Brain, Behavior, and Immunity 79 (2019): 63–74, 10.1016/j.bbi.2019.04.034. [DOI] [PubMed] [Google Scholar]
- 136. Winger R. C. and Zamvil S. S., “Your Nose Knows How to Target Brain Inflammation,” Brain 139, no. Pt 7 (2016): 1866–1869, 10.1093/brain/aww121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Howells D. W., Sena E. S., and Macleod M. R., “Bringing Rigour to Translational Medicine,” Nature Reviews. Neurology 10, no. 1 (2014): 37–43, 10.1038/nrneurol.2013.232. [DOI] [PubMed] [Google Scholar]
- 138. Head M. L., Holman L., Lanfear R., Kahn A. T., and Jennions M. D., “The Extent and Consequences of p‐Hacking in Science,” PLoS Biology 13, no. 3 (2015): e1002106, 10.1371/journal.pbio.1002106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Jha R. M., Rajasundaram D., Sneiderman C., et al., “A Single‐Cell Atlas Deconstructs Heterogeneity Across Multiple Models in Murine Traumatic Brain Injury and Identifies Novel Cell‐Specific Targets,” Neuron 112, no. 18 (2024): 3069–3088.e4, 10.1016/j.neuron.2024.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Koupourtidou C., Schwarz V., Aliee H., et al., “Shared Inflammatory Glial Cell Signature After Stab Wound Injury, Revealed by Spatial, Temporal, and Cell‐Type‐Specific Profiling of the Murine Cerebral Cortex,” Nature Communications 15, no. 1 (2024): 2866, 10.1038/s41467-024-46625-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Li Y., Ru X., Xu Y., et al., “Single‐Cell Sequencing Reveals Intracranial Microvasculature‐Derived CXCL12 Promotes CD8,” Journal of Neuroinflammation 22, no. 1 (2025): 116, 10.1186/s12974-025-03444-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Hwang J. R., Byeon Y., Kim D., and Park S. G., “Recent Insights of T Cell Receptor‐Mediated Signaling Pathways for T Cell Activation and Development,” Experimental & Molecular Medicine 52, no. 5 (2020): 750–761, 10.1038/s12276-020-0435-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Keck S., Schmaler M., Ganter S., et al., “Antigen Affinity and Antigen Dose Exert Distinct Influences on CD4 T‐Cell Differentiation,” Proceedings of the National Academy of Sciences of the United States of America 111, no. 41 (2014): 14852–14857, 10.1073/pnas.1403271111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Allison K. A., Sajti E., Collier J. G., et al., “Affinity and Dose of TCR Engagement Yield Proportional Enhancer and Gene Activity in CD4+ T Cells,” eLife 5 (2016): 5, 10.7554/eLife.10134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Solouki S., Huang W., Elmore J., Limper C., Huang F., and August A., “TCR Signal Strength and Antigen Affinity Regulate CD8+ Memory T Cells,” Journal of Immunology 205, no. 5 (2020): 1217–1227, 10.4049/jimmunol.1901167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Snook J. P., Kim C., and Williams M. A., “TCR Signal Strength Controls the Differentiation of CD4,” Science Immunology 3, no. 25 (2018): eaas9103, 10.1126/sciimmunol.aas9103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Meng H., Zhao H., Cao X., et al., “Double‐Negative T Cells Remarkably Promote Neuroinflammation After Ischemic Stroke,” Proceedings of the National Academy of Sciences of the United States of America 116, no. 12 (2019): 5558–5563, 10.1073/pnas.1814394116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Shi L., Sun Z., Su W., et al., “Treg Cell‐Derived Osteopontin Promotes Microglia‐Mediated White Matter Repair After Ischemic Stroke,” Immunity 54, no. 7 (2021): 1527–1542.e8, 10.1016/j.immuni.2021.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Stubbe T., Ebner F., Richter D., et al., “Regulatory T Cells Accumulate and Proliferate in the Ischemic Hemisphere for up to 30 Days After MCAO,” Journal of Cerebral Blood Flow and Metabolism 33, no. 1 (2013): 37–47, 10.1038/jcbfm.2012.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Kilgore M., “Created in BioRender,” (2025), https://BioRender.com/d08o727.
- 151. Kilgore M., “Created in BioRender,” (2025), https://BioRender.com/ail24za.
- 152. Kilgore M., “Created in BioRender,” (2025), https://BioRender.com/x97e653.
- 153. Kilgore M., “Created in BioRender,” (2025), https://BioRender.com/whafh18.
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
