Academic Editor

Article Metrics

  • Fig. 1.

    View in Article
    Full Image
  • Fig. 2.

    View in Article
    Full Image
  • Fig. 3.

    View in Article
    Full Image
  • Information

  • Download

  • Contents

Abstract

Neurotrauma plays a significant role in secondary injuries by intensifying the neuroinflammatory response in the brain. High Mobility Group Box-1 (HMGB1) protein is a crucial neuroinflammatory mediator involved in this process. Numerous studies have hypothesized about the underlying pathophysiology of HMGB1 and its role in cognition, but a definitive link has yet to be established. Elevated levels of HMGB1 in the hippocampus and serum have been associated with declines in cognitive performance, particularly in spatial memory and learning. This review also found that inhibiting HMGB1 can improve cognitive deficits following neurotrauma. Interestingly, HMGB1 levels are linked to the modulation of neuroplasticity and may offer neuroprotective effects in the later stages of neurotraumatic events. Consequently, administering HMGB1 during the acute phase may help reduce neuroinflammatory effects that lead to cognitive deficits in the later stages of neurotrauma. However, further research is needed to understand the time-dependent regulation of HMGB1 and the clinical implications of treatments targeting HMGB1 after neurotrauma.

1. Introduction

Neurotrauma refers to the sudden disruption of central nervous system (CNS) functioning or the development of new CNS pathology due to applied external forces [1, 2]. It represents a significant cause of morbidity and mortality worldwide, affecting both the young and the elderly. Two major events classified under neurotrauma include traumatic brain injury (TBI) and stroke. In the United States, TBI alone impacts approximately 2.8 million individuals annually [2]. Within the realm of trauma-related injuries, TBI is often referred to as a silent epidemic because the resulting impairments may not be immediately visible [2]. Previous studies have provided compelling evidence that individuals who survive a TBI may experience persistent consequences, including memory issues and neuropsychological challenges that initiate an ongoing, potentially lifelong process affecting multiple organ systems, possibly leading to the development or acceleration of various other neurological diseases [3], such as neurodegenerative diseases or epilepsy.

Stroke, on the other hand, is a global epidemic that affects 1 in 4 adults over the age of 25 which poses a significant individual, societal and economic burden as well [4]. Moreover, the study has shown that TBI can result in stroke, where any damage to the brain typically results in impairment to the vascular system. A stroke, caused by a disruption in the brain’s blood supply, is a cerebrovascular event resulting in the loss of brain function, thereby neurotrauma [4]. Therefore, it is reasonable to speculate that damage to the cerebrovascular system in the head caused by TBI could potentially increase the risk of stroke, either through bleeding from an artery (haemorrhagic stroke) or the formation of a clot at the site of injury that obstructs blood flow to the brain (ischemic stroke) [5, 6]. Cognitive impairments after neurotrauma poses a significant economic burden and leads to overall poor quality of life when patients are unable to return to work after the injury [7]. One study found high rates of morbidity and unemployment rates in patients who suffered a minor form of head injury 3 months post-trauma [8]. Approximately 30% of stroke patients have been found to develop dementia which mainly affects the domains of attention, memory, language and orientation within 1 year of the stroke onset [9]. Cognitive impairment may be the most significant comorbidity post neurotrauma as it affects a survivor’s quality of life, mainly in terms of their ability to carry out daily activities independently, readapting to social life, resumption of work and return to premorbid family roles [10].

Neuroinflammation occurs as a secondary response to neurotrauma which contributes to persistent and ongoing cognitive deficits and neurodegeneration [11]. This can be attributed to activation of microglia and astrocytes, leukocyte recruitment and migration via the blood-brain barrier (BBB) disruption and the increase in neurotoxic or neuroprotective inflammatory mediators [12]. Damage associated molecular patterns (DAMPs) released by injured neurons during neuroinflammation tend to interact with toll like receptors (TLRs) on activated microglial cells in order to induce the release of inflammatory mediators. These mediators in turn leads to further upregulation microglial activation, creating a self-perpetuating cycle that ultimately leads to chronically dysregulated microglial activation and the release of neurotoxic reactive oxygen species (ROS) in the brain, thus precipitating neurodegeneration [11, 13].

A key molecule that is involved in secondary insults to the brain following neurotrauma includes High Mobility Group Box-1 (HMGB1), a non- histone chromatin protein released by damaged cells which signals cellular damage and amplifies neuroinflammation [14]. Kang et al. [15] describes intranuclear HMGB1 as “a DNA chaperone which bends DNA and modulates crucial DNA events such as repair, replication, transcription, recombination and genomic stability”. HMGB1 that is secreted extracellularly binds to the receptor for advanced glycation end products (RAGE), which signals cell damage and acts as an inflammatory mediator. The ability of HMGB1 to be secreted out of the cell depends on whether the cell undergoes necrosis or programmed cell death [16]. HMGB1 is released from the nucleus and cytoplasm of the injured cell when cell lysis occurs. HMGB1 acts by binding to a variety of cell surface receptors to trigger immune cell chemotaxis and the release of proinflammatory cytokines. These surface receptors include RAGE, TLR2, TLR4 and TLR9 [17, 18, 19]. To date, the role of RAGE and TLR4 have been widely studied [20]. For example, the binding of HMGB1 to TLR4 further stimulates HMGB1 release. This signalling creates a self-perpetuating cycle, which further amplifies the neuroinflammatory response in the human brain [21].

HMGB1 binding to TLR4 also varies in necrotic and apoptotic cells. This can be attributed to its varied redox states. A reduced form of HMGB1 released in necrotic cells is required in order for it to activate TLR4, whereas HMGB1 released during programmed cell death is oxidised rendering it unable to bind with TLR4 signalling receptors. Hence inflammatory responses are only triggered in necrotic cells alone [22]. HMGB1 has been studied as a biomarker that can prognosticate TBI as evidenced by similarities in prognostic values of HMGB1 and Glasgow Coma Scale (GCS) [23]. There have been limited studies with regards to HMGB1 and cognition in the setting of overall neurotrauma. HMGB1 may be a promising target for neurotrauma initiated cognitive decline treatment in future studies. Therefore, this review aims to determine the role of HMGB1 in cognition in the setting of neurotrauma and to explore the potential for HMGB1 as a target for treatment and prevention of neurocognitive decline by summarizing and critically appraising the currently available literature on their pathological relationship.

2. Current Literature on HMGB1 in Neurotrauma Related Cognition

A total of 1854 articles were retrieved from the initial literature database search where a total of 1322 articles were excluded due to duplicates and non-original research articles. An additional 513 articles that did not discuss HMGB1 in relation to cognition as well as articles unrelated to the subject of neurotrauma related cognitive deficits were also excluded. The remaining 19 studies, summarized in Table 1 (Ref. [24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41]), were included for critical appraisal. All 19 articles were pre-clinical studies investigating the relationships between HMGB1 and cognitive deficits in the context of neurotrauma. These studies included 11 on traumatic brain injury (TBI), 6 on stroke (both ischemic and hemorrhagic), and 2 on hypoxia-induced neurotrauma.

Table 1. Table of results of HMGB1 findings and cognitive outcomes across various disease modalities, sample characteristics and mechanisms involved in the studies reviewed.
Type of Neurotrauma Sample Characteristics HMGB1 Findings Cognitive Findings Mechanisms Involved Study
TBI Inducible HMGB1 KO C57BL/6 mice & Age matched WT littermate M, (N = 45) Loss of HMGB1 was associated with significantly smaller lesion volume in HMGB1 KO mice. No difference in spatial learning in WT and HMGB1 KO mice after controlled cortical impact (CCI). RAGE and TLR4 Aneja et al. 2019 [24]
16–20 weeks old
TBI IL-1R KO B6.129S7-Il1r1tm1Imx & Age matched WT control mice, M, (N = 335) HMGB1 was released from contused brain tissue 24 h post CCI. IL-1R KO mice demonstrated decreased hippocampal learning and working memory post CHI. IL-1 signaling. Chung et al. 2019 [25]
3–6 months old No difference between HMGB1 levels in cortex, hippocampus, and striatum in sham and CHI mice at 24 h and 48 h.
HMGB1 translocation was increased in cortical and hippocampal regions in all CHI animals compared to sham animals post CHI.
TBI Adult SD rats & Sham control rats, M, (N = 44) A combination of TBI and alcohol led to a 5 fold increase of cortex HMGB1 expression when compared to either alcohol or TBI alone. Results showed that alcohol-exposed animals had decreased frequency of recognition to the novel object when compared to alcohol-naïve controls. TLR and RAGE Teng et al. 2015 [26]
8–10 weeks old It is speculated that this enhanced HMGB1 expression reflects accentuated neuroinflammation in animals exposed to chronic alcohol exposure after TBI.
TBI C57BL/6 mice, M (N = 130) Au -treated mice that underwent TBI had lower levels of HMGB1 than non-treated TBI mice. Au improved memory functions in mice that underwent TBI. TLR4 Wang et al. 2020 [27]
Adult Nrf2 knockdown significantly increased expression levels of HMGB1 as compared with those of the TBI + Au + negative control group.
TBI C57Bl/6 mice, M (N = 12–15 per group) Glycyrrhizin (Gly) treatment prior to injury, but not after injury, reduced acute HMGB1 levels. Gly treatment selectively prevented spatial and motor learning deficits post TBI. TLR4 Webster et al. 2019 [28]
3 weeks old Sham Gly and TBI Gly groups showed significantly higher cognitive performance compared to TBI Vehicle mice.
TBI Wistar rats, M, 2 groups, (N = 5) Plasma HMGB1 in rats that underwent injury was increased. Gly improved the memory during the subacute phase of TBI. RAGE Okuma et al. 2014 [29]
9–11 weeks old Gly inhibited HMGB1 translocation in neurons and the binding between HMGB1 and RAGE receptors.
TBI SD rats, M, (N = 51) Cytosolic HMGB1 was significantly increased at 24 h in injured animals versus control. Rats treated with minocycline showed improved spatial memory acquisition but there was no difference between sham versus controlled cortical impact (CCI) minocycline-treated rats on any days tested. NA Simon et al. 2018 [30]
Post-natal day 17 Treatment with minocycline significantly reduced cytosolic HMGB1 in injured rats.
No significant difference in cytosolic HMGB1 was seen between naïve and minocycline-treated injured rats.
TBI SD rats, M (N = 40) CCI increased the hippocampal HMGB1 expression. It was not significantly reduced by Nec-1 or melatonin administration. Nec-1 and melatonin improved learning and memory decline caused by CCI. RIP1/R IP3-M LKL signaling pathway Bao, et al. 2019 [31]
8 weeks old Cytoplasmic HMGB1 release in CCI group was increased. This was alleviated by Nec-1 and melatonin. A20 silencing aggravated the CCI-induced decrease in learning and spatial memory ability.
AAV-shA20 also significantly ameliorated the beneficial effects of Nec-1 and melatonin in decreasing CCI- induced cognitive and memory deficits.
TBI SD rats, Sex NA, (N = 80) HMGB1 was significantly increased in the cortex in the SAH group as compared to that of the control group. Following SAH, melatonin treatment significantly reduced spatial learning and memory deficits. TLR4 Wang et al. 2013 [32]
Adult HMGB1 in the brains of SAH+ melatonin group were significantly lower than those of the SAH vehicle group.
TBI SD rats, M, (N = 48) HMGB1 expression in the miR-216a-5p and BDNF-induced MSCs-Exo groups was significantly lower than that in the PBS group after TBI. BDNF-induced MSCs-Exo significantly promoted the recovery of spatial learning function in rats after TBI. NF-κB Xu et al. 2020 [33]
Adult
Stroke-Hemorrhagic C57BL/6 mice, M (N = 25) HMGB1 expression in the perihematomal and hippocampal brain region was significantly upregulated in the diabetes (Db), intracerebral hemorrhage (ICH) and diabetes/ICH (Db/ICH) groups compared with the sham group in the perihematomal and hippocampal brain regions. Db/ICH mice suffered from higher cognitive deficits compared to Db, ICH and sham groups in terms of spatial learning in T-maze test. RAGE and TLR4 Bahader et al. 2021 [34]
8–14 weeks old No statistical differences in the HMGB1 expressions were noted between these groups. No significant differences noted in cognitive deficits between Db and ICH groups.
Stroke-Ischemia SD rats, M, (N = 70) SSa inhibited nuclear HMGB1 of the MCAO rat brain. SSa attenuated the learning and memory recovery in the MCAO rat. TLR4/NF-κB signaling pathway Wang and Yang, 2020 [35]
Age NA SSa pre-treatment reduced serum HMGB1 during reperfusion.
Stroke-Ischemia ICR mice, M, (N = 32) Minocycline treatment reversed GCI- induced increases of HMGB1. Reduced learning and memory ability occurred in GCI. NA Du et al. 2019 [36]
Age NA Plasma HMGB1 was significantly reduced with Minocycline administration. No significant improvement in spatial and learning memory in the Minocycline group.
Stroke-Ischemia SD rats, M (N = 169) Anti-HMGB1 neutralising Ab decreased nuclear HMGB1 translocation in the hippocampus. Cognitive performance in the MWM was significantly improved at 4–12w in the anti-HMGB1 group compared to the PBS group. NA Hei et al. 2018 [37]
4–6 weeks old Hippocampal HMGB1 decreased significantly in the PBS group compared to the sham group.
HMGB1 neutralization preserved BBB integrity, and suppressed glial activation, proinflammatory cytokine production and oxidative stress in the acute phase of CCH.
These changes exerted long-time beneficial effects on hippocampal neuronal survival and cognitive function in the chronic phase.
Stroke-Ischemia SD rats, M, (N = 130 rats) Celastrol decreased tGCI/R induced HMGB1 upregulation and IκBα phosphorylation, and reversed the nuclear translocation of NF-κB. Celastrol (4 mg/kg) improve spatial learning and memory of tGCI/R rats vs other studies. NF-κB signaling pathway Zhang et al. 2020 [38]
Adults Therefore, neuroprotective effects of celastrol could be largely attributed to the suppression of inflammatory cascades via an HMGB1-dependent NF-κB signaling pathway.
Stroke-Ischemia SD rats, M, (N = 4/group) Ischemic stroke significantly decreased hippocampal HMGB1 and FC treatment inhibited this decrease. FC treatment improved the spatial learning ability in the MCAO rats. NA Zhang et al. 2019 [39]
8–10 weeks old
Hypoxia BALB/c mice, M, (N = 65) Mac-1 knockdown significantly decreased the cytoplasmic HMGB1 in both PFC and primary microglia culture. Mac-1 suppression decreased working memory loss. TLR4 Das et al. 2018 [40]
6–10 weeks old
Hypoxia ICR mice, M, (N = 143) HMGB1 mRNA level in the ipsilateral HI hippocampus of the HI + PBS group was significantly increased compared with that in the sham group while this elevated level was significantly decreased after Gly treatment. HI + PBS mice demonstrated significantly impaired spatial learning ability. TLR4, NF-κB and MyD88 pathway Pascual et al. 2011 [41]
Postnatal day 7 Medium and high doses of Gly improved learning and spatial memory impairment caused by HI insult.

Abbreviations: HMGB KO, High Mobility Group Box Knockout; WT, Wild Type; CCI, Controlled Cortical Impact; RAGE, receptor for advanced glycation end products; TLRs, toll-like receptors; TBI, Traumatic Brain Injury; CHI, Closed Head Injury; IL-1R KO, Interleukin-1 Receptor Knockout; Db, Diabetic; ICH, Intracranial Haemorrhage; Au, Aucubin; Nrf2, Nuclear factor erythroid-2 related factor 2; Gly, Glycyrrhizin; Nle4, D-Phe7, NDP-melanocortin analogue; A20, Tumor necrosis factor alpha induced protein 3; Nec-1, Necrostatin-1; SAH, Subarachnoid Haemorrhage; SD, Sprague Dawley; BDNF, Brain-Derived Neurotrophic Factor; MSCs-Exo, mesenchymal stem cell-derived exosome; PBS, Phosphate Buffered Saline; SSa, Saikosaponin A; MCAO, Middle Cerebral Artery Occlusion; tGCI/R, Transient Global Cerebral Ischemia Reperfusion; FC, Fluorocitrate; PFC, Prefrontal Cortex; HI, Hypoxia Ischemia; Ab, Antibody; NA, not available; AAV, adeno-associated virus; NF-κB, nuclear factor kappa B; CCH, chronic cerebral hypoperfusion.

The role of HMGB1 as an inflammatory biomarker in neurotrauma associated cognitive impairment was explored in the studies selected for this review. The studies cited explored the effects of therapeutics such as Melatonin, Nec-1, Aucubin, Thalidomide, Minocycline, Glycyrrhizin, Saikosaponin A, Necrostatin-1, melanocortin analogue and celastrol on hippocampal, cortical, serum and cerebrospinal serum fluid (CSF) HMGB1 levels. Subsequently, the effects of disease modalities such as hypoxia, stroke and traumatic brain injuries on spatial memory, working memory, spatial learning and recognition memory were explored. A total of 10 studies were conducted using Sprague Dawley rat models, 3 studies utilized of C57BL/6 mice, 1 study consisted of HMGB1-knockout (KO) transgenic mice, 1 study consisted of Wistar rats, 1 study consisted of interleukin (IL)-1 receptor KO transgenic mice, 1 study consisted of BALB/c mice and 2 studies consisted of ICR mice. Thirteen of these studies consisted of adult mice between 6 weeks to 24 months of age. Four studies consisted of younger mice of less than 6 weeks. The age of mice used in the remaining studies were unspecified. Male mice were used in 18 of the experimental models. The sex of subjects used in 1 study was unspecified.

The majority of non-treatment based studies showed increased HMGB1 upregulation and decreased cognitive performance of mice that experienced TBI, stroke and hypoxic injury. Hippocampal HMGB1 levels were reduced across all intervention groups studies. Subjects belonging to the treatment arm in the majority of these studies showed significant improvement in spatial learning and memory acquisition.

3. Discussion

Overall, the currently available literature provided clear evidence regarding the role of HMGB1 as a key mediator and link between neurotrauma and the consequent neuroinflammation that may have led to neurotrauma-related cognitive impairment. The studies (Table 1, Ref. [24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41]) demonstrated an increased HMGB1 expression within the hippocampus of rats postinjury and this was linked with an increased susceptibility to impairments in spatial learning and memory acquisition. Furthermore, the literature also showed that the suppression of HMGB1 ameliorated the cognitive deficits and that it may have potential for the development of targeted therapy to improve cognitive outcomes in a clinical setting of neurotrauma.

3.1 HMGB1 and its Potential Mechanisms Affecting Cognition
3.1.1 TLR4/TLR2/RAGE Mechanism

High Mobility Group Box-1 (HMGB1) is a protein that behaves as a nuclear factor that is secreted extracellularly by activated monocytes and macrophages during necrosis and cellular damage. Within the nucleus, it modifies the structure of the DNA via chromatin bending in order to initiate the assembly of DNA targets [15]. HMGB1 that is secreted extracellularly binds to the various receptors such as RAGE, TLR4 and TLR2 which signals cellular damage and behaves as an inflammatory mediator [17, 41, 42]. HMGB1 binding to these receptors leads to NF-κB upregulation which subsequently leads to the release of cytokines such as IL-1 and IL-6 [43, 44]. These cytokines then bind to cognate receptors present on nearby neuronal cells and this in turn upregulates intracellular signaling, and further amplifies the inflammatory cascade. This exacerbates chronic neuronal injury and death and subsequently leads to cognitive decline as shown in Fig. 1 [32].

Fig. 1.

A potential mechanism of HMGB-1 induced cognitive decline in neurotrauma via TLR4/TLR2/RAGE/NF-κB signalling. TNF, tumor necrosis factors; RAGE, receptor for advanced glycation end products; IL, interleukin; TLR, toll like receptors; TNFR1, tumor necrosis factor receptor 1.

3.1.2 Microglial and Astrocyte Activation

HMGB1 in various oxidative or reductive forms serve various purposes during the inflammatory cascade in the brain during an insult. Disulphide HMGB1-induced cytokine production mediates the activation of NF-κB signalling in microglial cells and macrophages which leads to N-methyl-d-aspartate (NMDA)-induced neuronal cell death in the hippocampus when bound to TLR-4 receptors. For example, HMGB1 in its disulfide form increases the influx of Ca2+ into neuronal cells which occurs as a result of NMDA activation which can lead to increased neuronal excitability and subsequent neuronal loss [45].

A study on the effect of Glycyrrhizin on HMGB1 showed that HMGB1 inhibition led to a reverse in hypoxic-ischemic-induced myelination disorder and hippocampal neuronal loss via the TLR4/RAGE/NF-κB pathway, which led to better cognitive outcomes. HMGB1 was postulated to mediate hippocampal neuronal damage by activating microglial and astrocytic cells [46]. Microglial cells cease to modulate plasticity once polarized by HMGB1. HMGB1 promotes the release of proinflammatory factors, leading to secondary neuronal damage [47]. On the other hand, the continuous release of HMGB1 leads to an excess astrocyte proliferation. Chronic astrocytic activation leads to impaired axonal regeneration and scar formation in regions of the brain that underwent neuronal loss. The hippocampal neuronal damage that follows microglial and astrocytic HMGB1 induced activation has been postulated to increase cognitive deficits [48] (Fig. 2).

Fig. 2.

A potential mechanism of HMGB-1 induced cognitive decline in neurotrauma via loss of healthy hippocampal neurons due to microglial and astrocyte activation.

3.1.3 Loss of Healthy Myelin

The loss of healthy myelin has been proposed to lead to cognitive decline. HMGB1 was postulated to increase inflammatory and oxidative stress upon oligodendrocytes, which play a role in myelin formation [49]. The loss of healthy myelin that follows could potentially lead to changes in neuronal firing, synaptic plasticity and Hebbian learning, which is a mechanism by which neurons become more efficient at communicating when they are repeatedly activated together. However, when myelin is damaged, this process can be disrupted, leading to further cognitive impairment [50, 51] (Fig. 3).

Fig. 3.

A potential mechanism of HMGB-1 induced cognitive decline in neurotrauma via loss of healthy myelin.

HMGB1 was postulated to increase blood brain barrier permeability in neurotrauma in many studies. Using HMGB1 KO mice and Wild Type (WT) mice, Aneja and colleagues [24] studied the effect of HMGB1 loss on tissue sparing and cognition post Controlled Cortical Impact (CCI) and described that naive HMGB1 KO mice after TBI exhibited no difference in performance in spatial memory testing compared to WT mice when cognitive improvement was expected in the absence of HMGB1 in the mice. Aneja and colleagues [24] postulated that these differences could be attributed to differences in species studied (rat model vs mouse model) and differences in models and severity of the injuries. Despite the absence of HMGB1, blood brain barrier permeability increased 24 h post injury, indicating that other DAMPs besides HMGB1 may have played a role in increasing BBB permeability [24].

BBB breakdown has been postulated to cause the increase of blood related immune cell chemotaxis into the brain that leads to activation of resident immune CNS cells and subsequent inflammatory amplification [52]. However, HMGB1 may not be the only DAMPs involved in mediating BBB breakdown. Chung and colleagues [25] and Teng and colleagues [26] found an increase in HMGB1 level post TBI in the setting of CCI or Closed Head Injury (CHI) and alcohol exposure post TBI respectively, both of which were associated with poorer cognitive outcomes. Teng and colleagues [26] speculated that the increase in HMGB1 expression could signify that neuroinflammatory processes were aggravated by alcohol ingestion post TBI via astroglial activation and enhanced release of proinflammatory cytokines, which ultimately led up to increased secondary injuries. Teng and colleagues [26] established a significantly positive correlation between HMGB1 and Neurological Severity Score (NSS), suggesting that HMGB1 release may have contributed to neuroinflammatory processes and further exacerbated neurocognitive decline.

Bahader and colleagues [34] proposed that ICH increased the induction of proinflammatory cells and mediators such as HMGB1, which in turn led to higher proportion of glial and astrocytic activation and an increase in oxidative and nitrosative stress, leading to raised hematoma volume and severe neurological and cognitive deficits via TLR4 and RAGE receptor activation [27, 53].

Thus, it is acknowledged that intracellular HMGB1 plays a role in repair and regulation of autophagy, whereas extracellular HMGB1 acts as a trigger and enhancer of neuroinflammation. The initiation of neuroinflammatory responses often stems from inherent “damaging” events. During neuroinflammation, HMGB1 is primarily secreted by activated microglial cells. This secretion induces heightened release of inflammatory factors such as tumor necrosis factors (TNF)-α, IL-1β, and IL-6 through activation of relevant inflammatory pathways, subsequently leading to neurological damage and dysfunction [54]. Following neurotrauma, several comorbid pathologies such as oxidative stress and mitochondrial dysfunction can significantly impact HMGB1 pathways and cognitive function [55]. However, the interactions of these pathologies on the role of HMGB1 in neuroinflammation and cognitive outcomes following neurotrauma remain to be elucidated, emphasizing the need for comprehensive management strategies targeting both HMGB1 pathways and associated comorbid pathologies.

3.2 Therapeutic Targets of HMGB1
3.2.1 In Traumatic Brain Injury

Wang and colleagues found that Aucubin (Au) treated mice had lower HMGB1 expression and improved cognitive outcomes when compared to mice that did not receive treatment. It was postulated that Au reduced oxidative stress by inhibiting intracellular reactive oxygen species (ROS) and increasing the concentration of serum and brain enzymes that inhibit ROS synthesis [27]. Au, by reducing oxidative stress in the hippocampus has a potential to reduce neuronal loss and alleviate cognitive deficits in secondary injuries induced by TBI. This suggests that HMGB1 may lead to downstream increase in oxidative stress which may exacerbate neuronal injury and death, leading to cognitive decline.

Simon and colleagues found that Minocycline’s ability to block the translocation of HMGB1 reduced microglial activation within the thalamus post TBI. This may have had a neuroprotective effect via inhibition of nuclear HMGB1 release and subsequent cytoplasmic HMGB1 release from the lysed injured cell leading to better cognitive outcomes in the treatment group, similar to that of Gly [30]. In another experiment conducted by Bao and colleagues, Nec-1 and Melatonin were found to reduce necroptosis in an A-20 (TNF) dependent manner [31]. In other words, the inhibition of HMGB1, RAGE, NF-κB pathways, cytokine release and subsequent cognitive competence was dependent on A-20 upregulation [56]. Wang Z and colleagues [32] demonstrated that melatonin plays an important role in inhibiting subarachnoid haemorrhage- induced TLR4- mediated agents (upstream agents: TLR4, MyD88, NF-κB and downstream agents: IL-1b, TNF-a, IL-6, and iNOS) [33].

TLR4 activation and subsequent direct NF-κB activation was MyD88 dependent (Kang and Lee [57] 2012) (Kang et al. [56] 2011). Due to inhibition of these pathways, and the reduction of neuroinflammation and improvement in cognitive outcomes that followed, it was suggested that melatonin was an effective anti-inflammatory agent in the setting of secondary injuries due to SAH. Xu and colleagues [33] had similar findings in terms of HMGB1 and cognition, whereby HMGB1 in miR-216a-50 and BDNF induced exosomes were lower that of the PBS group [34] miR-216a-50 was found to inhibit apoptosis by targeting the HMGB1/NF-κB pathways, leading to a subsequent reduction in neuroinflammation [56, 59]. BDNF, a neuroprotective protein, was postulated to have been related to miR-216a-50 and it’s downstream target protein, HMGB1 [34].

3.2.2 In Ischemia and Hypoxia Injury

Wang, X and colleagues found that Saikosaponin (SSa) pretreatment attenuated the increase in HMGB1 and nuclear NF-κB and was associated with neuroprotective effects and improvement in neurological recovery after ischemia and reperfusion injury [35]. Similarly, Celastrol inhibition of glial activation and of HMGB1/NF-κB pathways led to reduced apoptotic neuronal cell death, which was associated with better cognitive outcomes [38]. In a study conducted by Das and colleagues [40], Mac-1receptor suppression showed a reduction in TLR4 and NF-κB expression induced by hypoxia. It also resulted in the transcriptional upregulation of M2a, a neuroprotective phenotype of microglia. Suppression in neuroinflammatory mechanisms induced by HMGB1 was postulated to be associated with improvement in cognitive functioning. The study found that Mac-1 suppression induced a decrease in GSK3b which was associated with beta amyloid deposition, a pathophysiological feature which is known to induce cognitive decline in neurodegenerative diseases such as Alzheimer’s Disease [40].

Du and colleagues [36] also explored the effect of Minocycline but global cerebral ischemia (GCI). In this study, Minocycline (MIN) was noted to significantly reduce serum HMGB1 levels, the release of its downstream inflammatory mediators and subsequent microglial activation. However in this study, no significant cognitive improvement was observed, but a trend of cognitive improvement was seen in the group that received MIN [36]. This may have been attributed to cognitive function being studied in the short term similar to that of Okuma and colleagues [29].

3.3 HMGB1 Regulation Is A Time Dependent Regulation

Studies showed that Glycyrrhizin (Gly) treatment reduced the expression of HMGB1 and prevented and improved cognitive decline. Webster and colleagues found that Gly reduced brain HMGB1 treatment and reduced brain edema on the ipsilateral side of the injury, a hallmark feature of neuroinflammation [58, 59]. However, this finding was limited to mice receiving treatment prior to injury and within 1 hour after the injury, suggesting that Gly may have beneficial effects in TBI in a very acute setting [29]. Gly also was shown to have beneficial effects on cognition in terms of cognitive outcomes. The neuroprotective effects of Gly were hypothesized to be attributed to the downregulation of HMGB1 via TLR4 inhibition and the subsequent decline in hippocampal microglial activation [53]. Similar results were observed in a study conducted by Okuma and colleagues [29], who postulated Gly caused RAGE inhibition in an acute setting. In this experiment however, Gly’s beneficial effects ceased at most after 7 days. However, studies about cognition were not conducted after 7 days and therefore the long term effects of Gly were not established in this study and have not been investigated in previous literature as well.

In another experiment, Fluorocitrate (FC) aided stroke induced memory impairment via structural alterations in astroglial cells [39]. It was also noted that HMGB1 was raised in a biphasic manner (3 days and 2 weeks postoperatively) and that FC administration had to be done in a prudent manner as HMGB1 played the role in mediation of neuroplasticity and neuroinflammation depending on the stages of cerebral ischemia reperfusion injury [60]. Therefore, it was suggested that FC treatment was best avoided during the recovery phase of ischemic stroke, in order to promote HMGB1 mediated neurological remodelling [39]. Similarly, in another study, it was found that raised HMGB1 levels of hippocampal cornu ammonis (CA1) neurons occured 3 days after surgery could suggest that HMGB1 release occurred in the acute phases of chronic cerebral hypoperfusion (CCH) and that hippocampal HMGB1 likely originates from CA1 neurons. The study also found that remarkable neuronal loss and cognitive impairment occurred in the control group during the chronic phases of CCH, indicating that acute treatment of neurotrauma could potentially ameliorate cognitive deficits in the chronic phases of the disease [37]. This could potentially explain why across the majority of studies, targeted therapy in the acute phases of neurotrauma led to improvement of cognitive outcomes over the long term.

4. Conclusions

Based on these selected studies, HMGB1 appears to be a crucial mediator of the neuroinflammatory processes in the hippocampus that occur during/post- neurotrauma, which functions via the stimulation of TLR4/RAGE/NF-κB pathways. The subsequent loss of microglial and astrocytic inhibition within the hippocampus leads to reduced hippocampal neuronal loss and overall improvement in neurotrauma-related cognitive outcomes. Furthermore, based on several studies, it can be concluded that due to the biphasic nature of HMGB1 release in disease modalities such as strokes, treatment modalities targeting HMGB1 work effectively in the active phase of the disease. This could be attributed to the duality in the nature of HMGB1 whereby it functions as a neuroinflammatory mediator and modulator of neuroplasticity and neurological remodelling in active and chronic phases respectively. Therefore, it could be postulated that HMGB1 given at an acute phase may be beneficial in reducing neuroinflammatory effects which manifest as cognitive deficits in the later phases of neurotrauma. However, the mechanism of HMGB1 time dependent regulation warrants further studies.

Abbreviations

HMGB KO, High Mobility Group Box Knockout; WT, Wild Type; CCI, Controlled Cortical Impact; TBI, Traumatic Brain Injury; CHI, Closed Head Injury; IL-1R1 KO, Interleukin-1 Receptor Knockout; Db, Diabetic; ICH, Intracranial Haemorrhage; Au, Aucubin; Nrf2, Nuclear factor erythroid-2 related factor 2; Gly, Glycyrrhizin; Nle4, D-Phe7, NDP-melanocortin analogue; MSH, melanocyte-stimulating hormone; A20, Tumor necrosis factor, alpha induced protein 3, Nec-1, Necrostatin-1; SAH, Subarachnoid Haemorrhage; SD, Sprague Dawley; BDNF, Brain-Derived Neurotrophic Factor; MSCs-Exo, mesenchymal stem cell-derived exosomes; PBS, Phosphate Buffered Saline; SSa, Saikosaponin A; MCAO, Middle Cerebral Artery Occlusion; tGCI/R, Transient Global Cerebral Ischemia Reperfusion; FC, Fluorocitrate; PFC, Prefrontal Cortex; HI, Hypoxia Ischemia; Ab, Antibody.

Author Contributions

TR, AA and MFS have conceptualized and designed the study. LN performed the literature search and drafted the manuscript. TR and LN collected and assembled the data. LN, TR, AA and MFS analyzed the articles. TR, AA and MFS revised manuscript critically. All authors read and approved the final manuscript version. All authors contributed to editorial changes in the manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received funding from the Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Seed Grant 2022 and the Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Seed Grant 2023.

Conflict of Interest

The authors declare no conflict of interest. Given his role as a Guest Editor, Mohd. Farooq Shaikh had no involvement in the peer-review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to Thomas Heinbockel.

References

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.