Citation: Jian Zou, Fulton T. Crews. Glutamate/NMDA excitotoxicity and HMGB1/TLR4 neuroimmune toxicity converge as components of neurodegeneration[J]. AIMS Molecular Science, 2015, 2(2): 77-100. doi: 10.3934/molsci.2015.2.77
[1] | Edith Debroas, Carine Ali, Dominique Duval . Dexamethasone enhances glutamine synthetase activity and reduces N-methyl-D-aspartate neurotoxicity in mixed cultures of neurons and astrocytes. AIMS Molecular Science, 2015, 2(2): 175-189. doi: 10.3934/molsci.2015.2.175 |
[2] | Viktor Semenkov, Anatolii Mikhalskii, Alexandr Sapoznikov . Influence of heat shock proteins in individual sensitivity of human neutrophils to heat stress. AIMS Molecular Science, 2019, 6(2): 38-51. doi: 10.3934/molsci.2019.2.38 |
[3] | Flavia di Michele, Elena Romeo . What do we need to know about neurosteroids and emotions?. AIMS Molecular Science, 2023, 10(2): 70-78. doi: 10.3934/molsci.2023006 |
[4] | Seidu A. Richard, Wu Min, Zhaoliang Su, Hua-Xi Xu . Epochal neuroinflammatory role of high mobility group box 1 in central nervous system diseases. AIMS Molecular Science, 2017, 4(2): 185-218. doi: 10.3934/molsci.2017.2.185 |
[5] | William M. Scovell, Sachindra R. Joshi . The changing paradigm: estrogen receptor α recognition on DNA and within the dynamic nature of nucleosomes. AIMS Molecular Science, 2015, 2(2): 48-63. doi: 10.3934/molsci.2015.2.48 |
[6] | Morgan Robinson, Brenda Yasie Lee, Zoya Leonenko . Drugs and drug delivery systems targeting amyloid-β in Alzheimer's disease. AIMS Molecular Science, 2015, 2(3): 332-358. doi: 10.3934/molsci.2015.3.332 |
[7] | Paul Daniel Phillips, Timothy Andersen, Owen M. McDougal . Assessing the utility and limitations of high throughput virtual screening. AIMS Molecular Science, 2016, 3(2): 238-245. doi: 10.3934/molsci.2016.2.238 |
[8] | Thomas Rauen, Rose Tanui, Christof Grewer . Structural and functional dynamics of Excitatory Amino Acid Transporters (EAAT). AIMS Molecular Science, 2014, 1(3): 99-125. doi: 10.3934/molsci.2014.3.99 |
[9] | Marta Monzón . Approaches to therapy against prion diseases focused on the individual defence system. AIMS Molecular Science, 2017, 4(3): 241-251. doi: 10.3934/molsci.2017.3.241 |
[10] | Aliya I Sani, Zil-e-Rubab, Shumaila Usman, Syed Zaryab Ahmed, Mervyn Hosein . Role of OX40 and its ligand as costimulatory modulators in cancer immunotherapy. AIMS Molecular Science, 2021, 8(3): 161-173. doi: 10.3934/molsci.2021012 |
Neuronal degeneration and cell loss in brain are features of neurological disorders such as Parkinson's Disease (PD), Amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and Alzheimer's disease (AD) [1,2,3]. Although a great deal of effort has been devoted to explore the molecular and cellular mechanism responsible for the development and progression of these pathologies, the neuropathogenesis of the initiation or progression of neurodegeneration remains unclear. Neuroinflammation and excitotoxicity are considered to be distinct but interconnected mechanisms contributing to the neuropathogenesis of neurodegenerative disorders [4]. Excitotoxicity is caused by excessive activation of glutamate/ N-methyl-d-aspartate (NMDA) receptors due to accumulation of glutamate in extracellular space and is considered to be one of the main causes of neurodegeneration. Evidence of excessive glutamatergic signaling has been found in several neurodegenerative disorders [5]. In recent years, it has been postulated that neuroinflammation and excitotoxicity frequently coexist in neurological disorders. Many stud i es indicate that proinflammatory cytokines released from dying neurons activate microglia and contribute to the process of excitotoxicity through blocking glial glutamate transporters, releasing glial glutamate [4,6,7] and /or potentiating glutamate receptor function [8]. In general, neuroimmune activation has been considered to involve responses to dying cells inducing a wide variety of proinflammatory molecules that endanger neighboringcells in autocrine/paracrine fashions by spreading the proinflammatory signaling. In this manuscript we present data indicating that glutamate toxicity and HMGB1/TLR signaling in parallel contributes to neurotoxicity.
High mobility group box-1 (HMGB1), a ubiquitous nuclear and cytosolic non-histone DNA-binding protein, has multiple signaling mechanisms including activation of TLR4 receptors. HMGB1 can be released into extracellular space in one of two settings: active secretion or passive release from necrotic or damaged cells [9,10,11,12]. Extracellular HMGB1 functions as a damage associated molecular pattern (DAM P) or an alarmin [13] and activates proinflammatory signaling pathways by activating pattern recognition receptors including Toll-like receptor (TLR) 2 and 4 and receptor for advanced glycation end products (RAGE) [14]. HMGB1 has multiple forms related to oxidation state that have different activities at these receptors, with a disulfide-HMGB1 form having full TLR4 agonist activity, that is not a property of the reduced all thiol-HMGB1, although it retains cytokine-like activation of RAGE and other receptors, whereas fully oxidized HMGB1 is inactive [15,16]. Actively released HMGB1 is acetylated and disulfide-HMGB1, whereas cell death induced release from necrotic cells involves disulfide-HMGB1 and other active forms, whereas release from apoptotic cells is inactive HMGB1 [17]. Despite extensive studies in immunocompetent cells showing that HMGB1 is actively released into the extracellular space during sterile inflammation, few studies have investigated release of HMGB1 from neuronal cells. We and others find HMGB1 is abundantly expressed in neurons [11,18,19,20] including human cortical neurons [21]. HMGB1-TLR4 signaling is linked to epilepsy kindling of hyperexcitability and seizures [22]. Other studies have also reported that targeting HMGB1 can offer neuroprotection in animal models of brain injuries [23,24,25] and reverse memory impairments [26].
We used an ex vivo model of hippocampal-entorhinal cortex (HEC) slice cultures that contain all brain cell types and maintain the morphological structure of the intact brain to test the hypothesis that delayed neuronal death is due to convergence of glutamate-excitotoxic and HMGB1/TLR signaling that ultimately lead to neuronal death. We report here that HMGB1 is released by glutamate/NMDA or mGluR agonists and that neuroimmune signaling induced neuronal death involves glutamate/NMDA receptors. HMGB1 release precedes neuronal cell death induced by glutamate/NMDA as well as by neuroimmune signaling activated by proinflammatory cytokine TNFα and endotoxin lipopolysaccharide (LPS). Furthermore, antagonists to glutamate/NMDA receptors or HMGB1 block neuronal death independent of the initial triggering stimulus. Although glutamate antagonists protect against neurotoxicity, HMGB1 antagonists may allow protection without directly disrupting normal glutamate synaptic signaling, a critical component of normal brain function.
The following reagents were purchased from Sigma-Aldrich (St Louis, USA) : Glutamic acid, NMDA, LPS from Escherichia coli 0111:B4, glycyrrhizin, ifenprodil and HMGB1. TNFα was purchased from R&D System (Minneapolis, USA). Non oxidizable chemokine-HMGB1 and cytokine-HMGB1 came from HMG Biotech (Milano, Italy). Lipopolysaccharide from the photosynthetic bacteriumRhodobacter sphaeroides (Lps-Rs) came from InvivoGen (San Diego, USA). HMGB1 ELISA kit and neutralizing antibodies were purchased from IBL International (Hamburg, Germany).
All protocols followed in this study were approved by the Institutional Animal Care Use Committee at UNC and were in accordance with National Institute of Health regulation for the care and use of animal in research. The preparation of organotypic hippocampal-entorhinal cortex (HEC) slice cultures was described elsewhere [8]. Briefly, hippocampal-entorhinal cortex complexes were dissected from neonates on postnatal day 7 in Guey’s buffer and transversely sectioned at thickness of 375 µm. Individual HEC slice was placed onto tissue insert membrane (10‒12 slices/insert) and cultured with medium containing 75% MEM with 25 mM HEPES and Hank’s salts + 25% horse serum (HS) + 5.5 g/L glucose + 2 mM L-glutamine in a humidified 5% CO2 incubator at 36.5 °C for 7 days in vitro (DIV), followed by 4 DIV in medium containing 12.5% HS and then 3 DIV in serum-free medium supplemented with N2. The cultures after 14 DIV were used for experiments and drug treatments with serum-free N2-supplemented medium.
All drug treatments were initiated on slices at 14DIV in serum-free N2-supplemented medium. For dose-response studies, HEC slices were treated with various concentrations of drugs (see below) for a fixed time (24 or 48 hrs), and then slices and media were removed for further analysis. In time-course studies, HEC slices were treated with a fixed concentration of drug depending on the experimental design (see below) and a total of 100 µL media was removed at each time point from culture for ELISA measurements of HMGB1. After removing media, the same amount of 100 µL fresh N2 medium was added into culture.
Media HMGB1 levels were determined with ELISA kit (IBL, Germany) according to the manufacturer’s instruction. A total of 50 µL of culture medium from each sample was used for ELISA. All samples were run in triplicate.
The uptake of the fluorescent exclusion dye propidium iodide (PI) was used for determination of neuronal cell death. PI is a polar compound that is impermeable to a cell with an intact cell membrane but penetrates damaged cell membranes. Inside the cells it binds nuclear DNA to generate the brightly red fluorescence. This method has been well characterized as accurately measuring neuronal degeneration in organotypic slice cultures [8,27,28] and the PI uptake was confirmed to significantly correlate with LDH efflux [28,29], which is another well accepted marker for neuronal cell death [30]. For each experiment, PI was added into the culture medium at the beginning of treatment at a concentration of 5 mg/mL and PI fluorescence images were captured at indicated time points. PI fluorescent intensity was measur ed and analyzed with the AxioVision 3.1 software. Using an interactive drawing tool, hippocampal CA1 field was outlined as regions of interest (ROIs), and mean PI fluorescent intensity of ROI was then determined by the program [31].
HEC slices at 14 DIV were treated with NMDA (200 µM) for 40 min. After NMDA treatment, NMDA-containing medium was removed from cultures and replaced with fresh serum-free N2-supplemented medium. After 24 hrs, culture media were collected and pooled as conditioned medium (CM). Pooled CM was diluted (1:3) and then used for treatment of new slice cultures.
HEC slice cultures were removed at the end of the experiment and fixed with 4% paraformaldehyde + 5% sucrose in 0.01 M PBS for 24 hrs at 4 °C. Free-floating slices were used for immunofluorescent staining. For detection of HMGB1 in PI-labeled neuronal cells, PI-labeled (red) slices after images captured for measurements of neuronal cell death were directly fixed and removed for performing immunofluorescent staining of rabbit anti-HMGB1 (1:500, AbCam). For double immunofluorescent staining, HEC slices were stained with neuronal marker MAP2 (mouse, 1:1000, sigma) and HMGB1 (Rabbit, 1:500, AbCam). All primary antibodies were incubated for 48 hrs at 4 °C. Either Alexa Fluor 594 or Alexa Fluor 488 secondary antibodies (1:2000; Molecular Probes, Eugene, OR) were used for immunofluorescent staining and incubated for 1 hr at room temperature. The slices were coverslipped with anti-fade mounting medium (pro-long; Molecular Probes). Confocal analysis was performed using a LeicaSP2 AOBS Upright Laser Scanning Confocal in Michael Hooker Microscopy Facility (University of North Carolina, Chapel Hill, NC).
Cell lysates were prepared from the entire HEC slices. Briefly, HEC slices were incubated in RIPA lysis buffer plus phosphatase and /or protease inhibitor cocktail (Thermo Scientific, USA) for 15 min and then disrupted with sonication (3 time, 2 min apart in ice). After centrifugation of the slice homogenate, the supernatant were collected, and protein level determined using a BioRad Bradford reagent kit (BioRad). For Western blotting, an equal amount of protein (50 μg) was mixed with 10 μL 5× loading buffer, and separated using a 4‒15% Tris mini-gel (Bio-Rad) and transferred onto PVDF membrane. After blocking with LI-COR blocking buffer overnight, the membrane was probed with mouse anti-phospho-NR2B (1:1000, Santa Cruz), rabbit anti-total NR2B antibodies (1:200, Santa Cruz) and mouse anti-β-actin (1:1000, Santa Cruz) at 4 °C overnight. After washing, membrane was incubated with second antibodies coupled with fluorescence from LI-COR Bioscience and then scanned with Odyssey machine (Lincoln, NE).
Data are expressed as a mean values ± st and ard error of mean from the indicated number of slices or experiments. Statistical comparisons were made with ANOVA and the difference between the experimental groups was further compared by using post hoc Fisher PLSD test. Differences were considered to be statistically significant if p value of < 0.05.
Hippocampal-entorhinal cortex (HEC) brain slice cultures provide an ex vivo model validated by many to investigate mechanisms of neurotoxicity [8,27,28]. In the present study, we tested the hypothesis that HMGB1 contributes to glutamate/NMDA receptor excitotoxicity. The glutamate/NMDA concentration-dependence of HMGB1 release into the media is compared to neuronal death as indexed by measurements of PI intensity within hippocampal CA1 field of HEC slice. Glutamate at lower concentrations triggered robust release of HMGB1 while neuronal cell death shows no significant increase (Figure 1). Glutamate released HMGB1 by 50%, 350%, and 700% with 100 µM, 500 µM, and 1 mM, concentrations which show no increase in neurotoxicity (Figure 1). Treatment of HEC slices with glutamate at concentration of 5 mM increased media HMGB1 by 1700%, accompanying with a marked 300% increase in neuronal death measured as PI fluorescent intensity (Figure 1A-dash line, 1B-c). Similarly, t reatment of HEC slices with various concentration of NMDA for 24 hrs drastically increased media HMGB1 level by 500‒800% at concentrations up to 1 µM, all concentrations showing no significant increase in neuronal death (Figure 2). NMDA at 10 µM and 100 µM increased media HMGB1 by 1400% and 2000% and significantly increased neuronal cell death by 350% and 650% respectively (Figure 2A-dash line, 2B-c). Immunoprecipitation of media proteins using an antibody for acetyl-lysine followed by western blot using an antibody for HMGB1 found that glutamate and NMDA increased release of acetyl-HMGB1, a marker of actively released HMGB1 [11]. We also tested the effects of stimulating mGluR on secretion of HMGB1. We treated HEC slices with mGluRII agonist LY-354740 (100 nM) or non-selective mGluR agonist (1S, 3R) -1-aminocyclopentane-1, 3-dicarboxylic acid (ACPD, 100 µM). Both LY-354740 and ACPD stimulated significant increases in media HMGB1 levels (Control: 1.8 ± 0.2; LY-354740: 11.2 ± 0.4; ACPD: 14.2 ± 2.6, p < 0.001, n = 3) without causing significant neuronal death (PI Intensity: Control 24.4 ± 3.5; LY-354740 32.1 ± 6.9; ACPD 26.7 ± 5.9, p < 0.09, n = 6‒8). LY-354740 and ACPD-stimulated HMGB1 release was blocked by mGluR1/II antagonists LY3578 and MCPG (data not shown). Together, these results indicate that glutamate receptor activation can release HMGB1 through mGluR receptors that are not neurotoxic and through NMDA receptors at concentrations below those that trigger neurotoxicity. In addition, neurotoxicity further increases HMGB1 release.
Time course studies were conducted to correlate glutamate/NMDA stimulated release of HMGB1 with neuronal cell death. In this set of experiments, HEC slices were treated with neurotoxic concentrations of glutamate (3 mM) or NMDA (10 µM). Glutamate stimulated a rapid increase in media HMGB1 with 3 and 4 fold increases at 4 and 8 hrs time points when there was no measurable neuronal cell death (Figure 3). Similarly, NMDA treatment increases media HMGB1 after 4 hrs of treatment, without causing measurable neuronal death. Treatment for longer time periods by glutamate (24 and 48 hrs) or NMDA (8, 24 and 48 hrs) increased neuronal cell death and induced greater increases in media HMGB1 (Figure 3). To visualize cellular HMGB1, we performed double immunofluorescent staining and confocal image analysis from slice culture treated with NMDA (100 µM). The representative confocal images are depicted showing nuclear HMGB1 in neuronal cells at zero time (T0), with increased depletion of nuclear HMGB1 and cytoplasmic accumulation in neuronal cells 30 min (Figure 3E, b), and 2 hrs after NMDA (Figure 3E, c), early time points when there is no neuronal death. Treatment with NMDA for 8 and 24 hrs finds PI-labeled dying neurons (dense red nuclei) with little nuclear HMGB1 (Figure 3E, d, e, f). Higher magnification of confocal images after NMDA treatment for 24 hrs results in granule-like HMGB1+ staining that is confined within cytoplasm and proximal neuronal processes (Figure 3E, f). These findings suggest that stimulated HMGB1 release precedes neuronal death by excitotoxic concentrations of glutamate and NMDA and that neuronal death further increases HMGB1 release.
To further investigate the effects of glutamate on release of HMGB1 into the media, we determined the effect of glutamate receptor antagonists. MK-801 (25 µM) completely prevented glutamate and NMDA induced neuronal cell death. Glutamate and NMDA increased media HMGB1 from a control value of 2.6 ± 0.2 ng/mL (Figure 4A-B) to 18 ± 0.6 ng/mL and 53 ± 2.0 ng/mL for glutamate and NMDA respectively. MK801 reduced media HMGB1 to 3.7 ± 0.6 ng/mL and 4.3 ± 0.8 ng/mL in Glutamate-MK801 and NMDA-MK801 treated slices respectively. Confocal analysis of HMGB1-immunofluorescent staining found that nuclear HMGB1 was reduced in the nuclei of neurons treated with NMDA (Figure 4C-e). Controls and NMDA/MK-801 groups show prominent nuclear HMGB1 suggesting blockade of NMDA receptors blocks HMGB1 translocation to the cytoplasm and release of HMGB1 (Figure 4C-d, f). We also examined the effects of glutamate-AMPA receptor antagonist NBQX. Treatment with AMPA antagonist NBQX (30 µM) also reduced glutamate (1 and 3 mM, data not shown) stimulated HMGB1 release. These findings indicate that MK-801 blocks glutamate and NMDA stimulated HMGB1 release and neurotoxicity.
To investigate the direct impact of HMGB1 on glutamate/NMDA excitotoxicity, we conducted experiments by adding exogenous HMGB1 to HEC slice cultures during treatments of glutamate or NMDA. From a separate experiment testing the effects of exogenous HMGB1 on neuronal cell viability, HMGB1 alone for 48 hrs caused modest increases in PI intensity at concentrations less than 500 ng/mL, but clearly increased cell death at 500 ng/mL and 1000 ng/mL. The PI intensity values for HMGB1 alone were Control: 17.5 ± 1.8; HMGB1 100 ng/mL: 22.3 ± 2.6, p > 0.5 compared to control; HMGB1 500 ng/mL: 32.6 ± 3.2, p < 0.01 compared to Control; HMGB1 1000 ng/mL: 41.5 ± 6.2, p < 0.0003 compared to Control; n = 6‒8). These are high concentrations, which may be related to the commercial preparation of HMGB1 which is known to vary across lots from different vendors (11) or due to some loss of HMGB1-TLR4 activation due to HMGB1 inactivated by oxidation. HMGB1 (100 ng/mL) significantly and synergistically increased neuronal cell death when combined with glutamate (1 mM) or NMDA (10 µM) for 24 hrs (Figure 5A). Furthermore, we prepared conditioned media (CM) collected from NMDA-stimulated slice cultures, in which HEC slice cultures were pretreated with NMDA (200 µM) for 40 min and then replaced with fresh N2-suplemented medium to remove NMDA and capture released HMGB1 and other factors for 24 hrs (see method), and treated HEC slice with CM during glutamate/NMDA treatments. Combined treatments of HEC slices with glutamate (1 mM) or NMDA (10 µM) for 46 hrs significantly potentiated neuronal cell death by 103% and 49% respectively (Figure 5B). Addition of anti-HMGB1 neutralizing antibodies or HMGB1 inhibitor glycyrrhizin blocked CM-potentiated glutamate/NMDA neurotoxicity (Figure 5B). Taken together, these results indicate that HMGB1 con tributes to glutamate/NMDA neurotoxicity and that HMGB1 released from NMDA excited neurons can increase excitotoxic cell death.
To facilitate our underst and ing of the role of HMGB1 in excitotoxic delayed neuronal cell death induced by glutamate/NMDA, we performed experiments targeting HMGB1 by adding HMGB1 neutralizing antibodies or the HMGB1 inhibitor glycyrrhizin into HEC slice cultures during glutamate/NMDA treatment. Glutamate (1 mM) increases media HMGB1 by about 5, 8 and 9 fold at 8, 24 and 48 hrs, respectively (Figure 6A) consistent with glutamate releasing HMGB1 from cells that contributes to glutamate excitotoxicity. Glutamate stimulated HMGB1 release was significantly reduced by glycyrrhizin (G+Gly) likely due to blocking HMGB1 activation of TLR4 receptors that further stimulates release of HMGB1 [11]. Assessment of neuronal cell death indicated that glutamate treatment for 8 hrs does not show an increase in cell death, but at 24 and 48 hrs neuronal cell death is progressively increased (Figure 6A). Glutamate stimulated cell death was reduced by both HMGB1 neutralizing antibodies and by the HMGB1 antagonist glycyrrhizin (Figure 6B). Similarly, treatment with the receptor specific agonist, NMDA (10 µM), increased media HMGB1 level by about 7, 9 and 10 fold at 8, 24 and 48 hrs time points respectively, which corresponds to 220%, 400% and 310% increases of PI fluorescent intensity, e.g. neuronal cell death at each time point (Figure 6C‒D). The addition of HMGB1 neutralizing antibodies and inhibitor glycyrrhizin during NMDA treatment significantly reduced neuronal cell death (Figure 6C‒D). Together, these results provide evidence that HMGB1 released by glutamate receptor activation can contribute to glutamate/NMDA neurotoxicity.
To facilitate our underst and ing whether HMGB1 contributes to neuronal cell death caused by neuroimmune action, we investigated HMGB1 release and neuronal cell death induced by endotoxin LPS, the prototype TLR4 agonist, and the proinflammatory cytokine TNFα that we have previously found to increase sensitivity to glutamate neurotoxicity [31]. We first performed dose-response curve experiments, in which HEC slices were treated with different concentrations of LPS (25, 50 and
100 ng/mL) or TNFα (25, 50 and 100 ng/mL) for 48 hrs and media HMGB1 determined. The results indicate that media HMGB1 was increased by around 13, 14 and 16 fold with LPS and by 7, 10 and 11 fold with TNFα at concentrations tested respectively (data not shown). Time course determinations with LPS (50 ng/mL) and TNFα (50 ng/mL) find rapid increases in media HMGB1, with a 6 and 4 fold increase by LPS and TNFα respectively after treatment for 2 hrs (not shown). At the 8 and 24 hrs time points, media HMGB1 levels were 8 and 12 fold increases with LPS and 6 and 9 fold increases with TNFα relative to corresponding control level at each time point (Figure 7). All of these increases in HMGB1 release occur without a significant change in neuronal cell death relative to controls (Figure 7). However, after 48 hrs a significant increase in neuronal cell death is associated with further increases in media HMGB1 levels (Figure 7). The presence of HMGB1 neutralizing antibodies or inhibitor glycyrrhizin did not prevent HMGB1 release in response to LPS and TNFα, but it did reduce neuronal cell death induced by LPS and TNFα at the 48 hrs time point (data not shown). Co-treatment of LPS and TNFα with minocycline, an inhibitor of microglial cell activation, reduced both the release of HMGB1 and neuronal cell death (Figure 8), suggesting that microglia contribute to HMGB1 signaling and delayed neuronal cell death. Taken together, these results suggest that active release of HMGB1 precedes neuronal cell death in response to LPS and TNFα that contributes to neurotoxicity.
It has been demonstrated that HMGB1-TLR4 signaling sensitizes glutamate-NMDA receptors through NR2B containing NMDA receptors [22,32]. We further confirmed that blockade of all NMDA receptors with MK801, blocked both LPS and TNFα induced neuronal death, but not HMGB1 release into the media (Figure 9). The subunits of NMDA receptors contribute to altered glutamate sensitivity as well as neuronal localization. The NR2B subunit is particularly associated with neurotoxicity perhaps due to an extra-synaptic localization on neurons. We tested the hypothesis that NR2B may be involved in LPS and TNFα induced neurotoxicity using ifenprodil, a selective blocker of NR2B-containing NMDA receptors [33]. Ifenprodil blocked neuronal cell death caused by LPS and TNFα, but appears not to effect media HMGB1 release (Figure 9). These findings indicate that neuroimmune agonists stimulate HMGB1 release independent of glutamate-NMDA NR2B receptor induced neurotoxicity. Further, they indicate neuroimmune agonists increase glutamate/NMDA receptor stimulation, likely through glutamate release, contributing to neuroimmune agonist neurotoxicity.
To further underst and the role of NR2B, we performed Western blot analysis. Treatments with HMGB1 and LPS increased protein level of phosphorylated NR2B (Figure 9C). LPS- and TNFα-induced expression of NR2B phosphorylation was reduced by HMGB1 antagonist glycyrrhizin (Figure 9C). Taken together, these results indicate that HMGB1 modulation of NR2B is involved in delayed neuronal cell death induced by LPS and TNFα.
It has been found that the redox state of HMGB1 alters signaling with only disulfide-HMGB1 being an agonist at TLR4 [32]. To investigate the form of HMGB1 involved in excitotoxicity we used chemically modified HMGB1; non-oxidizable HMGB1 (Ox-HMGB1) with a triple cysteine-to-serine replacement (3S-HMGB1) which is a RAGE agonist, but not a TLR4 agonist, and the disulfide-containing form of HMGB1 (Dis-HMGB1), which is a TLR4 agonist with cytokine activity. HEC slices were treated with glutamate alone or in combination with Ox-HMGB1 or Dis-HMGB1 and the results indicate that glutamate (1 mM) neurotoxicity is potentiated by Dis-HMGB1, with a 32% and 82% increase at concentrations of 5 and 10 ug/mL, respectively (Figure 10A). Non-oxidizable HMGB1 at 5 µM has no effect on glutamate-induced neurotoxicity, but at 10 µM results in a 32% decrease in glutamate neurotoxicity (Figure 10A). We evaluated the effects of TLR4 antagonist Lps-Rs on HMGB1 potentiation of glutamate-induced neurotoxicity. We co-treated HEC slices with TLR4 antagonist Lps-Rs (pretreatment for 2 hrs) during HMGB1-glutamate treatment. HMGB1 (100 ng/mL) potentiated glutamate (1 mM) -induced neuronal death by 76% and TLR4 antagonist Lps-Rs blocked HMGB1 potentiation of glutamate neurotoxicity (Figure 10B). Together, these results suggest that ac tivation of TLR4 receptors plays a prominent role in mediating HMGB1 potentiation of glutamate neurotoxicity.
We report here that glutamate/NMDA excitotoxicity and neuroimmune toxicity share common elements that contribute to neurodegeneration. Glutamate and NMDA concentration response curves and time courses find release of HMGB1 at lower concentrations and earlier time points than those inducing neuronal death. For example, glutamate/NMDA treatment for 4hrs finds media HMGB1 increased 6‒7 fold with no apparent neuronal cell death. Further, treatment of slices for 48 hrs with concentrations of glutamate < 1 mM and NMDA <1 µM increase HMGB1 release into the culture media many fold without causing measurable neuronal death. Neuronal death was associated with massive release of HMGB1 at excitotoxic concentrations of glutamate/NMDA. Further, we found glutamate/NMDA stimulation increased media acetyl-HMGB1, consistent with activ e neuronal release of HMGB1 [11]. Our data strongly suggest that glutamate stimulated HMGB1 release precedes neuronal death. These findings are consistent with glutamate stimulated HMGB1 release contributing to delayed neuronal cell death induced by glutamate/NMDA excitotoxicity. Other studies find glutamate stimulation of neuronal firing releases HMGB1 [19] and increases NMDA stimulated neuronal calcium influx [29], the key signal mediating excitotoxicity [35]. Further, previous studies in primary neuronal cultures have found HMGB1 can mediate neuronal cell death through apoptosis [34] and potentiate NMDA-mediated necrotic cell death [32]. We found HMGB1 neutralizing antibodies and the inhibitor glycyrrhizin as well as blocking TLR4 activation all reduce glutamate/NMDA induced cell death. T hese findings support an important role for neuronal excitation releasing and increasing extracellular HMGB1 that potentiates excitotoxic neuronal death.
Neuroimmune gene expression is associated with most neurodegenerative diseases [35]. We report here that LPS and TNFα stimulation of neuronal death is reduced by ifenprodil and MK801, glutamate/NMDA receptor antagonists, consistent with glutamate release from neurons in response to LPS-TLR4 receptor activation contributing to the neuronal death. Release of HMGB1 leads to activation of TLR4 receptors and subsequently triggers more release of HMGB1. We found HMGB1 release increased phosphorylation of NR2B, an NMDA receptor subunit, that is associated with increased NMDA receptor signaling and increased NMDA mediated calcium flux [29]. The finding that blocking glutamate/NMDA signaling reduces LPS-TLR4 and TNFα toxicity indicates both processes contribute to neuronal death triggered through the respective signaling mechanisms. HMGB1-TLR4 signaling increases excitability through phosphorylation of glutamate/NMDA receptor NR2B, that increases excitability and risks of seizures [19,29] and glutamate release increasing excitotoxicity. These studies are consistent with HMGB1 and neuroimmune signaling increasing glutamate release and sensitivity to glutamate, with both signaling processes contributing to neuronal cell death.
Consistent with several previous studies [11,22,36], our results indicate that HMGB1 is predominantly released from neuronal cells in response to glutamate/NMDA and neuroimmune activation, though microglia and /or astrocytes may also be involved. Rapid release of HMGB1 from neuronal cells has been reported within 1 hr after the onset of middle cerebral artery occlusion in animal models of brain ischemia [37,38] and media HMGB1 increased 3 hrs following NMDA stimulation in the primary cultures of neuronal cells [34]. Consistent with these reports, the present study finds that the direct stimulation of glutamate receptors with glutamate, NMDA or mGluR agonists triggers rapid nucleocytoplasmic translocation and subsequent release of neuronal HMGB1, with 3 fold increase of media HMGB1 at 30min after applying neurotoxic concentration of NMDA (Figur e 3). In general, at 4 hrs treatments with sub toxic concentrations of glutamate/NMDA, media HMGB1 has increased 6‒7 fold while neuronal cell death has not been evident, whereas massive release of HMGB1 occurs with neuronal cell death. Our data strongly suggest that nuclear HMGB1 release proceeds neuronal death. Excitotoxicity is a leading cause of neuronal cell death in acute or chronic neurological conditions. Under pathological conditions, excessive activation of glutamate receptors causes Ca2+ influx and subsequent excitotoxicity [39]. Putative blockade of glutamate receptors in order to prevent neuronal cell death in neurodegenerative disorders has been disappointing in clinical trials due in part to the complex nature of excitotoxicity-induced neuronal death. The danger molecule-like actions of extracellular HMGB1 contributes stimulating glutamate release, which in turn increases neuronal toxicity [19]. In primary culture of neuronal cells, HMGB1 directly potentiates NMDA-stimulated calcium influx [32]. Animal models find that extracellular HMGB1 can worsen ischemic neurodegeneration [18]. Direct injection of HMGB1 into the hippocampus prior to injection of kainic acid significantly enhances the severity of seizures [22]. On the other h and , many studies have shown that targeting HMGB1 offers effective results in reducing neuronal death under neuropathological conditions. In animal models of ischemia, antagonizing HMGB1 by using HMGB1 neutralizing antibodies, antagonists or siRNA confer significant neuroprotection [23,40,41]. The present study further supports the notion that extracellular HMGB1 contributes to delayed neuronal death induced by excitotoxicity. Release of HMGB1 precedes neuronal cell death in response to glutamate/NMDA stimulation. Enhanced neuronal death can be effectively blocked by application of HMGB1 neutralizing antibodies and inhibitor glycyrrhizin. Ou r data in concert with others strongly suggest that HMGB1 may be mediating components of neuronal death. It is conceivable that underlying initial insults to brain such as inflammation, trauma and ischemia can cause release of glutamate and the latter in turn triggers mobilization of nuclear HMGB1 and subsequent release. To our knowledge, the present study is the first to show that blockade of glutamate receptor activation with antagonists including MK-801 and NBQX as well as mGluR antagonists prevent HMGB1 release into culture medium and reduce neuronal death. Confocal analysis indicates that MK-801 maintains nuclear localization of HMGB1 blocks HMGB1 release and neuronal cell death induced by glutamate/NMDA. These results suggest the possibility that stabilizing nuclear HMGB1 during brain insults may protect neuronal cells from excitotoxicity.
HMGB1-TLR4 signaling has been implicated in mediating neuroimmune responses under many neuropathological conditions inducing brain ischemia [42,43], post-traumatic brain edema [44] and ethanol-induced neuroinflammation [11]. Recent studies indicate that activation of TLR4 by HMGB1 is related to HMGB1 redox state. TLR4-mediated NMDA-induced Ca2+ influx and excitotoxicity are potentiated only by the disulfide-containing form of HMGB1, but not by non-oxidizable HMGB1 [32]. The present study further supports that HMGB1-TLR4 signaling contributes to glutamate/NMDA excitotoxicity, which is blocked by selective TLR4 antagonist Lps-Rs. We did not try to distinguish necrotic or apoptotic cell death, excitotoxicity often involves necrosis or a broader spectrum, e.g. necroptosis, consistent with release of TLR4 active HMGB1 contributing to glutamate necrotic neuronal death [16,29], however, actively released HMGB1 has been reported to induce neuronal apoptosis through RAGE activation [31]. RAGE activation by HMGB1 can occur with a partially oxidized HMGB1that does not have TLR4 activity [17]. We found that disulfide- HMGB1, the TLR4 agonist form of HMGB1, but not oxidized HMGB1 that is inactive at TLR4 and RAGE receptors, p otentiates glutamate/NMDA-induced neuronal cell death. Studies have found that HMGB1 contributes to neuronal death in models of ischemic-stroke [33,34,38], hemorrhagic stroke [37] and traumatic brain injury [40]. We found that Lps-Rs, the TLR4 antagonist, blocks glutamate/HMGB1 toxicity consistent with activation of TLR4 receptors contributing to excitotoxicity. Although we cannot rule out a contribution of HMGB1-RAGE signaling, these results suggest blocking HMGB1-TLR4 signaling increases excessive glutamate/NMDA receptor activation and that blocking HMGB1/TLR4 signaling could protect against neurodegeneration.
We further extend the study into the role of HMGB1 in neuroimmune signaling induced neurodegeneration. These studies indicate that neuroimmune signaling is a component of glutamate/NMDA mediated excitotoxic neuronal death. Treatments of endotoxin LPS and cytokine TNFα induce delayed (after 48hrs) neuronal cell death in HEC slices, which is enhanced by HMGB1. Time courses indicate that media HMGB1 is rapidly elevated in response to LPS and TNFα stimulation. At the 8 hrs time point, media HMGB1 levels were 10 and 7 fold increase with LPS and TNFα stimulation respectively while index of neuronal cell death is not increased (Figure 9). Media HMGB1 levels after 48 hrs correlated with increased neuronal death (Figure 9). Acetyl-HMGB1 release is increased indicating at least a portion of the HMGB1 release appears to be active release from excited neurons (These results suggest that active release of HMGB1 proceeds neuronal cell death induced by LPS and TNFα. HMGB1 likely increases NR2B-containing NMDA receptors increasing excitability [45]by enhancing NMDA function [32]. NMDA antagonists MK-801 and selective NR2B antagonist ifenprodil significantly reduced neuronal cell death induced by LPS and TNFα, suggesting glutamate increased activation of NR2B may be the molecular target modulated by HMGB1 to potentiate LPS and TNFα neurotoxicity through increased glutamate excitation. Interestingly the presence of ifenprodil has no inhibitory effects on media HMGB1 release while neuronal death is abrogated. This may be interpreted as the release of neuronal HMGB1 persists throughout treatment with LPS and TNFα involving distinct mechanisms independent of neuronal excitation and glutamate stimulation. These findings suggest HMGB1-TLR4 signaling and glutamate-NMDA signaling share common pathways to neuronal death.
In conclusion, the present study identifies danger signal HMGB1 as a molecule mediating delayed neuronal cell death induced by glutamate/NMDA as well as by LPS and TNFα. HMGB1 activation of TLR4 or modulation of NR2B represents molecular targets involving in HMGB1-mediated neuronal cell death. Targeting HMGB1 and its molecular targets may offer effective approaches to prevent neuronal death under neurological conditions.
This work was supported in part by the National Institutes of Health, National Institute on Alcoholism and Alcohol Abuse (AA019767, AA11605, AA007573, and AA021040), the Neurobiology of Adolescent Drinking in Adulthood (NADIA [AA020023, AA020024, and AA020022]), and the Bowles Center for Alcohol Studies. The authors wish to acknowledge support from the Bowles Center for Alcohol Studies and Michael Hooker Microscopy Facility, The University of North Carolina at Chapel Hill, School of Medicine. The authors would also like to thank Drs. Michael Chua and Neal Kramarcy for their technical support in confocal analysis and to Diana Lotito for assisting with the manuscript preparation. The authors declare no competing financial interests. All authors declare no conflict of interest in this paper.
All authors declare no conflicts of interest in this paper.
[1] |
Mattson MP (2000) Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol 1: 120-129. doi: 10.1038/35040009
![]() |
[2] |
Bredesen DE, Rao RV, Mehlen P (2006) Cell death in the nervous system. Nature 443: 796-802. doi: 10.1038/nature05293
![]() |
[3] |
Qin L, Wu X, Block ML, et al. (2007) Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 55: 453-462. doi: 10.1002/glia.20467
![]() |
[4] |
Tilleux S, Hermans E (2007) Neuroinflammation and regulation of glial glutamate uptake in neurological disorders. J Neurosci Res 85: 2059-2070. doi: 10.1002/jnr.21325
![]() |
[5] |
Sheldon AL, Robinson MB (2007) The role of glutamate transporters in neurodegenerative diseases and potential opportunities for intervention. Neurochem Int 51: 333-355. doi: 10.1016/j.neuint.2007.03.012
![]() |
[6] | Kostic M, Zivkovic N, Stojanovic I (2013) Multiple sclerosis and glutamate excitotoxicity. Rev Neurosci 24: 71-88. |
[7] |
Morimoto K, Murasugi T, Oda T (2002) Acute neuroinflammation exacerbates excitotoxicity in rat hippocampus in vivo. Exp Neurol 177: 95-104. doi: 10.1006/exnr.2002.7991
![]() |
[8] |
Zou JY, Crews FT (2005) TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition. Brain Res 1034: 11-24. doi: 10.1016/j.brainres.2004.11.014
![]() |
[9] |
Scaffidi P, Misteli T, Bianchi ME (2002) Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418: 191-195. doi: 10.1038/nature00858
![]() |
[10] |
Bell CW, Jiang W, Reich CF, et al. (2006) The extracellular release of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol 291: C1318-1325. doi: 10.1152/ajpcell.00616.2005
![]() |
[11] |
Zou JY, Crews FT (2014) Release of neuronal HMGB1 by Ethanol through decreased HDAC activity activates brain neuroimmune signaling. PloS One 9: e87915. doi: 10.1371/journal.pone.0087915
![]() |
[12] |
Evankovich J, Cho SW, Zhang R, et al. (2010) High mobility group box 1 release from hepatocytes during ischemia and reperfusion injury is mediated by decreased histone deacetylase activity. J Biol Chem 285: 39888-39897. doi: 10.1074/jbc.M110.128348
![]() |
[13] |
Lotze MT, Tracey KJ (2005) High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 5: 331-342. doi: 10.1038/nri1594
![]() |
[14] |
Andersson U, Tracey KJ (2011) HMGB1 is a therapeutic target for sterile inflammation and infection. Annu Rev Immunol 29: 139-162. doi: 10.1146/annurev-immunol-030409-101323
![]() |
[15] | Tang D, Billiar TR, Lotze MT (2012) A Janus tale of two active high mobility group box 1 (HMGB1) redox states. Mol Med 18: 1360-1362. |
[16] |
Janko C, Filipovic M, Munoz LE, et al. (2014) Redox modulation of HMGB1-related signaling. Antioxid Redox Signal 20: 1075-1085. doi: 10.1089/ars.2013.5179
![]() |
[17] | Magna M, Pisetsky DS (2014) The role of HMGB1 in the pathogenesis of inflammatory and autoimmune diseases. Mol Med 20: 138-146. |
[18] |
Faraco G, Fossati S, Bianchi ME, et al. (2007) High mobility group box 1 protein is released by neural cells upon different stresses and worsens ischemic neurodegeneration in vitro and in vivo. J Neurochem 103: 590-603. doi: 10.1111/j.1471-4159.2007.04788.x
![]() |
[19] |
Kim JB, Sig Choi J, Yu YM, et al. (2006) HMGB1, a novel cytokine-like mediator linking acute neuronal death and delayed neuroinflammation in the postischemic brain. J Neurosci 26: 6413-6421. doi: 10.1523/JNEUROSCI.3815-05.2006
![]() |
[20] |
Lo Coco D, Veglianese P, Allievi E, et al. (2007) Distribution and cellular localization of high mobility group box protein 1 (HMGB1) in the spinal cord of a transgenic mouse model of ALS. Neurosci Lett 412: 73-77. doi: 10.1016/j.neulet.2006.10.063
![]() |
[21] | Crews FT, Qin L, Sheedy D, et al. (2012) High Mobility Group Box 1/Toll-like Receptor Danger Signaling Increases Brain Neuroimmune Activation in Alcohol Dependence. Biol Psychiatry 73: 602-612. |
[22] |
Maroso M, Balosso S, Ravizza T, et al. (2010) Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat Med 16: 413-419. doi: 10.1038/nm.2127
![]() |
[23] |
Kim SW, Jin Y, Shin JH, et al. (2012) Glycyrrhizic acid affords robust neuroprotection in the postischemic brain via anti-inflammatory effect by inhibiting HMGB1 phosphorylation and secretion. Neurobiol Dis 46: 147-156. doi: 10.1016/j.nbd.2011.12.056
![]() |
[24] |
Ohnishi M, Katsuki H, Fukutomi C, et al. (2011) HMGB1 inhibitor glycyrrhizin attenuates intracerebral hemorrhage-induced injury in rats. Neuropharmacology 61: 975-980. doi: 10.1016/j.neuropharm.2011.06.026
![]() |
[25] |
Okuma Y, Liu K, Wake H, et al. (2014) Glycyrrhizin inhibits traumatic brain injury by reducing HMGB1-RAGE interaction. Neuropharmacology 85: 18-26. doi: 10.1016/j.neuropharm.2014.05.007
![]() |
[26] | Chavan SS, Huerta PT, Robbiati S, et al. (2012) HMGB1 mediates cognitive impairment in sepsis survivors. Molecular medicine 18: 930-937. |
[27] |
Brana C, Benham C, Sundstrom L (2002) A method for characterising cell death in vitro by combining propidium iodide staining with immunohistochemistry. Brain Res Brain Res Protoc 10: 109-114. doi: 10.1016/S1385-299X(02)00201-5
![]() |
[28] |
Noraberg J, Kristensen BW, Zimmer J (1999) Markers for neuronal degeneration in organotypic slice cultures. Brain Res Brain Res Protoc 3: 278-290. doi: 10.1016/S1385-299X(98)00050-6
![]() |
[29] |
Zimmer J, Kristensen BW, Jakobsen B, et al. (2000) Excitatory amino acid neurotoxicity and modulation of glutamate receptor expression in organotypic brain slice cultures. Amino Acids 19: 7-21. doi: 10.1007/s007260070029
![]() |
[30] |
Koh JY, Choi DW (1987) Quantitative determination of glutamate mediated cortical neuronal injury in cell culture by lactate dehydrogenase efflux assay. J Neurosci Methods 20: 83-90. doi: 10.1016/0165-0270(87)90041-0
![]() |
[31] |
Zou JY, Crews FT (2005) TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition. Brain Res 1034: 11-24. doi: 10.1016/j.brainres.2004.11.014
![]() |
[32] |
Balosso S, Liu J, Bianchi ME, et al. (2014) Disulfide-Containing High Mobility Group Box-1 Promotes N-Methyl-d-Aspartate Receptor Function and Excitotoxicity by Activating Toll-Like Receptor 4-Dependent Signaling in Hippocampal Neurons. Antioxid Redox Signal 21: 1726-1740. doi: 10.1089/ars.2013.5349
![]() |
[33] | Chenard BL, Menniti FS (1999) Antagonists selective for NMDA receptors containing the NR2B subunit. Curr Pharma Des 5: 381-404. |
[34] |
Kim SW, Lim CM, Kim JB, et al. (2011) Extracellular HMGB1 released by NMDA treatment confers neuronal apoptosis via RAGE-p38 MAPK/ERK signaling pathway. Neurotox Res 20: 159-169. doi: 10.1007/s12640-010-9231-x
![]() |
[35] |
Glass CK, Saijo K, Winner B, et al. (2010) Mechanisms underlying inflammation in neurodegeneration. Cell 140: 918-934. doi: 10.1016/j.cell.2010.02.016
![]() |
[36] |
Crews FT, Qin L, Sheedy D, et al. (2013) High mobility group box 1/Toll-like receptor danger signaling increases brain neuroimmune activation in alcohol dependence. Biol Psychiatry 73: 602-612. doi: 10.1016/j.biopsych.2012.09.030
![]() |
[37] |
Kim JB, Lim CM, Yu YM, et al. (2008) Induction and subcellular localization of high-mobility group box-1 (HMGB1) in the postischemic rat brain. J Neurosci Res 86: 1125-1131. doi: 10.1002/jnr.21555
![]() |
[38] |
Qiu J, Nishimura M, Wang Y, et al. (2008) Early release of HMGB-1 from neurons after the onset of brain ischemia. J Cereb Blood Flow Metab 28: 927-938. doi: 10.1038/sj.jcbfm.9600582
![]() |
[39] | Choi DW (1987) Ionic dependence of glutamate neurotoxicity. J Neurosci 7: 369-379. |
[40] |
Kim ID, Shin JH, Lee HK, et al. (2012) Intranasal delivery of HMGB1-binding heptamer peptide confers a robust neuroprotection in the postischemic brain. Neurosci Lett 525: 179-183. doi: 10.1016/j.neulet.2012.07.040
![]() |
[41] |
Sun Q, Wang F, Li W, et al. (2013) Glycyrrhizic acid confers neuroprotection after subarachnoid hemorrhage via inhibition of high mobility group box-1 protein: a hypothesis for novel therapy of subarachnoid hemorrhage. Med Hypotheses 81: 681-685. doi: 10.1016/j.mehy.2013.07.026
![]() |
[42] |
Zhang J, Wu Y, Weng Z, et al. (2014) Glycyrrhizin protects brain against ischemia-reperfusion injury in mice through HMGB1-TLR4-IL-17A signaling pathway. Brain Res 1582: 176-186. doi: 10.1016/j.brainres.2014.07.002
![]() |
[43] | Yang QW, Xiang J, Zhou Y, et al. (2010) Targeting HMGB1/TLR4 signaling as a novel approach to treatment of cerebral ischemia. Front Biosci 2: 1081-1091. |
[44] |
Laird MD, Shields JS, Sukumari-Ramesh S, et al. (2014) High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia 62: 26-38. doi: 10.1002/glia.22581
![]() |
[45] |
Maroso M, Balosso S, Ravizza T, et al. (2010) Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat Med 16: 413-419. doi: 10.1038/nm.2127
![]() |
1. | Manoj Banjara, Chaitali Ghosh, Sterile Neuroinflammation and Strategies for Therapeutic Intervention, 2017, 2017, 2090-8040, 1, 10.1155/2017/8385961 | |
2. | Anna Tyrtyshnaia, Anatoly Bondar, Sophia Konovalova, Ruslan Sultanov, Igor Manzhulo, N-Docosahexanoylethanolamine Reduces Microglial Activation and Improves Hippocampal Plasticity in a Murine Model of Neuroinflammation, 2020, 21, 1422-0067, 9703, 10.3390/ijms21249703 | |
3. | H. S. Swartzwelder, Kati L. Healey, Wen Liu, Kira Dubester, Kelsey M. Miller, Fulton T. Crews, Changes in Neuroimmune and Neuronal Death Markers after Adolescent Alcohol Exposure in Rats are Reversed by Donepezil, 2019, 9, 2045-2322, 10.1038/s41598-019-47039-1 | |
4. | Yi-Shu Huang, Joy Ogbechi, Felix I. Clanchy, Richard O. Williams, Trevor W. Stone, IDO and Kynurenine Metabolites in Peripheral and CNS Disorders, 2020, 11, 1664-3224, 10.3389/fimmu.2020.00388 | |
5. | Leonarda Di Candia, Edith Gomez, Emilie Venereau, Latifa Chachi, Davinder Kaur, Marco E. Bianchi, R.A. John Challiss, Christopher E. Brightling, Ruth M. Saunders, HMGB1 is upregulated in the airways in asthma and potentiates airway smooth muscle contraction via TLR4, 2017, 140, 00916749, 584, 10.1016/j.jaci.2016.11.049 | |
6. | Laura Medina-Ceja, Juan C. Salazar-Sánchez, Jorge Ortega-Ibarra, Alberto Morales-Villagrán, Connexins-Based Hemichannels/Channels and Their Relationship with Inflammation, Seizures and Epilepsy, 2019, 20, 1422-0067, 5976, 10.3390/ijms20235976 | |
7. | Gunnar R. Leitner, Tyler J. Wenzel, Nick Marshall, Ellen J. Gates, Andis Klegeris, Targeting toll-like receptor 4 to modulate neuroinflammation in central nervous system disorders, 2019, 23, 1472-8222, 865, 10.1080/14728222.2019.1676416 | |
8. | Kathryn A. Martinello, Christopher Meehan, Adnan Avdic-Belltheus, Ingran Lingam, Sara Ragab, Mariya Hristova, Cally J. Tann, Donald Peebles, Henrik Hagberg, Tim G. A. M. Wolfs, Nigel Klein, Ilias Tachtsidis, Xavier Golay, Boris W. Kramer, Bobbi Fleiss, Pierre Gressens, Nicola J. Robertson, Acute LPS sensitization and continuous infusion exacerbates hypoxic brain injury in a piglet model of neonatal encephalopathy, 2019, 9, 2045-2322, 10.1038/s41598-019-46488-y | |
9. | Chia-Ho Lin, Han-Yu Chen, Kai-Che Wei, Role of HMGB1/TLR4 Axis in Ischemia/Reperfusion-Impaired Extracellular Glutamate Clearance in Primary Astrocytes, 2020, 9, 2073-4409, 2585, 10.3390/cells9122585 | |
10. | Liya Qin, Jian Zou, Alexandra Barnett, Ryan P. Vetreno, Fulton T. Crews, Leon G. Coleman, TRAIL Mediates Neuronal Death in AUD: A Link between Neuroinflammation and Neurodegeneration, 2021, 22, 1422-0067, 2547, 10.3390/ijms22052547 | |
11. | Leon G. Coleman, Jian Zou, Fulton T. Crews, Microglial depletion and repopulation in brain slice culture normalizes sensitized proinflammatory signaling, 2020, 17, 1742-2094, 10.1186/s12974-019-1678-y | |
12. | Fawaz Alasmari, Hasan Alhaddad, Woonyen Wong, Richard L. Bell, Youssef Sari, Ampicillin/Sulbactam Treatment Modulates NMDA Receptor NR2B Subunit and Attenuates Neuroinflammation and Alcohol Intake in Male High Alcohol Drinking Rats, 2020, 10, 2218-273X, 1030, 10.3390/biom10071030 | |
13. | Leon G. Coleman, Jian Zou, Fulton T. Crews, Microglial-derived miRNA let-7 and HMGB1 contribute to ethanol-induced neurotoxicity via TLR7, 2017, 14, 1742-2094, 10.1186/s12974-017-0799-4 | |
14. | Fulton T. Crews, Ryan P. Vetreno, Cholinergic REST-G9a gene repression through HMGB1-TLR4 neuroimmune signaling regulates basal forebrain cholinergic neuron phenotype, 2022, 15, 1662-5099, 10.3389/fnmol.2022.992627 | |
15. | Fulton T. Crews, Leon G. Coleman, Victoria A. Macht, Ryan P. Vetreno, Robert Dantzer, Targeting Persistent Changes in Neuroimmune and Epigenetic Signaling in Adolescent Drinking to Treat Alcohol Use Disorder in Adulthood, 2023, 75, 0031-6997, 380, 10.1124/pharmrev.122.000710 | |
16. | Ningning Zhang, Hong Jiang, Huiqin Wang, Yating Wang, Ye Peng, Yangbo Liu, Congyuan Xia, Xu Yan, Shifeng Chu, Yi Zhang, Zhenzhen Wang, Naihong Chen, Novel Antidepressant Mechanism of Ginsenoside Rg1 in Regulating the Dysfunction of the Glutamatergic System in Astrocytes, 2022, 24, 1422-0067, 575, 10.3390/ijms24010575 | |
17. | Fulton T. Crews, Rachael P. Fisher, Liya Qin, Ryan P. Vetreno, HMGB1 neuroimmune signaling and REST-G9a gene repression contribute to ethanol-induced reversible suppression of the cholinergic neuron phenotype, 2023, 1359-4184, 10.1038/s41380-023-02160-6 | |
18. | Fulton T. Crews, Victoria Macht, Ryan P. Vetreno, Epigenetic regulation of microglia and neurons by proinflammatory signaling following adolescent intermittent ethanol (AIE) exposure and in human AUD, 2024, 4, 2674-0001, 10.3389/adar.2024.12094 | |
19. | Kaitlin R. Carroll, Mark Mizrachi, Sean Simmons, Bahtiyar Toz, Czeslawa Kowal, Jeffrey Wingard, Nazila Tehrani, Aida Zarfeshani, Nina Kello, Lara El Khoury, Rachel Weissman-Tsukamoto, Joshua Z. Levin, Bruce T. Volpe, Betty Diamond, Lupus autoantibodies initiate neuroinflammation sustained by continuous HMGB1:RAGE signaling and reversed by increased LAIR-1 expression, 2024, 1529-2908, 10.1038/s41590-024-01772-6 | |
20. | Shelby Travaglianti, Ahmed Alotaibi, Woonyen Wong, Magid Abou-Gharbia, Wayne Childers, Youssef Sari, Effects of novel GLT-1 modulator, MC-100093, on neuroinflammatory and neurotrophic biomarkers in mesocorticolimbic brain regions of male alcohol preferring rats exposed chronically to ethanol, 2024, 211, 03619230, 110935, 10.1016/j.brainresbull.2024.110935 | |
21. | Jian Zou, Elizabeth McNair, Sagan DeCastro, Scott P. Lyons, Angie Mordant, Laura E. Herring, Ryan P. Vetreno, Leon G. Coleman Jr, Microglia either promote or restrain TRAIL-mediated excitotoxicity caused by Aβ1−42 oligomers, 2024, 21, 1742-2094, 10.1186/s12974-024-03208-2 | |
22. | Mansi Dahalia, Sparsh Gupta, Haya Majid, Divya Vohora, , Pirfenidone regulates seizures through the HMGB1/TLR4 axis to improve cognitive functions and modulate oxidative stress and neurotransmitters in PTZ-induced kindling in mice, 2025, 15, 1663-9812, 10.3389/fphar.2024.1528032 |