Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2001 word(s) 2001 2021-03-08 06:37:30 |
2 update layout and reference Meta information modification 2001 2021-03-23 10:17:47 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Balança, B. Acute Phase of Brain Injury. Encyclopedia. Available online: https://encyclopedia.pub/entry/8150 (accessed on 04 July 2024).
Balança B. Acute Phase of Brain Injury. Encyclopedia. Available at: https://encyclopedia.pub/entry/8150. Accessed July 04, 2024.
Balança, Baptiste. "Acute Phase of Brain Injury" Encyclopedia, https://encyclopedia.pub/entry/8150 (accessed July 04, 2024).
Balança, B. (2021, March 21). Acute Phase of Brain Injury. In Encyclopedia. https://encyclopedia.pub/entry/8150
Balança, Baptiste. "Acute Phase of Brain Injury." Encyclopedia. Web. 21 March, 2021.
Acute Phase of Brain Injury
Edit

Early or primary injury due to brain aggression, such as mechanical trauma, hemorrhage or is-chemia, triggers the release of damage-associated molecular patterns (DAMPs) in the extracellular space. Some DAMPs, such as S100B, participate in the regulation of cell growth and survival but may also trigger cellular damage as their concentration increases in the extracellular space. When DAMPs bind to pattern-recognition receptors, such as the receptor of advanced glycation end-products (RAGE), they lead to non-infectious inflammation that will contribute to necrotic cell clearance but may also worsen brain injury.

acute brain injuries damage-associated molecular pattern molecules receptor for advanced glycation end-products biomarkers

1. Introduction

The cells of the innate immune system search the extracellular environment for exogenous pathogens or self-molecules—either modified (e.g., oxidized lipids) or usually confined within cells via pattern recognition and scavenger receptors (PRRs) [1][2][3]. In the central nervous system these receptors are mainly expressed by microglial cells but also by astrocytes, neurons, endothelial cells, and infiltrating leukocytes upon vessel injury or blood–brain barrier (BBB) opening [1][3][4]. Nevertheless, resident microglial cells have a larger repertoire of Toll-like receptors (TLRs) and a greater amount of the receptor for advanced glycation end-products (RAGE) [1][5][6]. According to the context, PRRs may polarize resident microglial cells and infiltrating leukocytes toward a panel of pro- or anti-inflammatory phenotypes, playing a role in tissue damage clearance and repair but which may also trigger neuronal and glial degeneration [1][3][5][7][8]. Although acute brain injuries encompass a wide variety of mechanisms, they share a common feature as they all lead to acute cellular necrosis with the release of intracellular ions, adenosine di- or triphosphate (ATP), proteins, and nucleic acids in the extracellular space, and eventually the extravasation of red blood cells and hemoglobin [9][10][11][12]. Collectively, this wide variety of endogenous molecules released upon tissue injury are termed damage or danger-associated molecular patterns (DAMPs).

Neurological recovery following acute brain injury depends on the patient’s prior medical condition and primary injuries, but also on secondary damage [13][14]. Among other secondary insults, the inflammation that is triggered by the release of DAMPs plays a crucial role in the development of brain lesions which will be discussed in the present review. The levels of inflammatory markers released in the systemic blood, such as the C-reactive protein, or interleukin (IL) 6 and 12, are actually predictors of post-stroke neurological dysfunction [14][15][16]. Moreover, the concomitant occurrence of a systemic inflammation, such as during sceptic or anaphylactic shock, increases neurological damage at the acute phase of stroke [17].

The acute increase of extracellular potassium after ischemia or hemolysis leads to the depolarization and swelling of neighboring cells thereby starting propagating waves of spreading depolarization [18] (Figure 1A,B). Potassium and ATP release from dying cells also activate the inflammasome in neurons and astrocytes via pannexin1, purinoreceptor, and nucleotide oligomerization domain receptors (NOD-like receptors) [1][19]. Larger molecules, such as S100 proteins, hemoglobin derivatives or the high mobility group protein 1 (HMGB1), bind mainly to TLRs and RAGE [1][10]; the ligation of TLRs and RAGE triggers a series of cellular signaling events, including the activation of nuclear factor-kappa B (NF-κB), leading to the production of pro-inflammatory cytokines, and causing non-sceptic inflammation [1][20][21]. However, the consequences of DAMPs ligation to PRRs depends on the context and their concentration in the extracellular space. For instance, S100B, a calcium binding protein that has several intracellular actions in astrocytes, can promote cell growth in the nM extracellular range (i.e., physiological conditions) [22][23][24]; whereas at higher concentration S100B activates astrocytes with a pro-inflammatory phenotype and facilitate neuronal death [24][25]. HMGB1 expression may also promote neuroinflammation related to brain injury but the deletion of the encoding gene is not able to prevent such consequences [26][27]. Furthermore, sustained activation and up-regulation of RAGE on neurons has been reported to cause death via stimulating the production of reactive oxygen species [28], but also regulates neurite growth and cell survival [24], as well as apoptosis and autophagy [20][29][30]. However, some extracellular soluble truncated receptors, such as sRAGE, act as decoys for ligands, and thus have a cytoprotective effect against advanced glycation end-products (AGE) and RAGE interactions; serum levels of sRAGE have been investigated in pathological process and proposed as a biomarker of their intensity and severity of outcome [31]. It seems that the net effect of DAMPs and PRRs, such as RAGE, depends on the context, cell type, and the number of DAMPs and the level of expression of PRRs.

Figure 1. Damage-associated molecular patterns (DAMPs) and pattern-recognition receptors (PRRs) changes at the acute phase of brain injury. (A) Morphological changes of neurons (grey), astrocytes (green), microglial and infiltrating leukocytes (blue), and DAMPs release (orange); (B) local field potential (LFP) and extracellular KCl recordings during the spreading depolarization triggered by the primary injury: the direct current (DC; 0–0.5Hz) shift is associated with a decrease of neuronal activity (AC; >0.5 Hz) and a KCl release; (C) Pattern-recognition receptors (PRRs) activation pathways; (D) kinetics of DAMPs and PRR expression as well as the course of cell death mechanisms. DAMPs: Damage-associated molecular patterns; IL: Interleukin; iNOS: inducible nitric oxide synthase; LFP: local field potential; MLKL: Mixed-lineage kinase domain-like pseudokinase; MyD88: Myeloid differentiation primary response 88; NFκB: nuclear factor-kappa B; RAGE: Receptor for advanced glycation end-products; RIPK1: receptor-interacting protein kinase 1; TLR: Toll-like receptor; TNF: Tumor necrosis factor; TNFR1: Tumor necrosis factor receptor 1.

2. Acute Lesion Progression Pathophysiology

2.1. Kinetics of DAMPs and Consequences after the Primary Insult

In addition to mechanical cell destruction, necroptosis is the main cell death pathway at the acute phase of traumatic brain injury (TBI), intracerebral or subarachnoid hemorrhage (SAH), and ischemic stroke (IS) [9][20][32]. Necroptosis is a morphologically lytic form of cell death implicating the receptor-interacting protein kinase 1 and 3 (RIPK1-RIPK3) and the mixed-lineage kinase domain-like pseudokinase (MLKL) pathway, resulting in the release of the contents of the cell into the extracellular space [33] (Figure 1A). The subsequent release of DAMPs peaks around 24 h after the primary insult and decreases thereafter over several days [4][12][20][34][35][36][37][38]. There is a spillover of DAMPs into the core of the primary injury, with a drastic decrease of intracellular HMGB1 [37] while HMGB1 is translocated from the nucleus to the cytoplasm of neurons but not glial cells at the periphery [4][9][37][39]. This extracellular release of HMGB1 may also act as a chemoattractant to monocytes [40] that infiltrate the core of the lesion from the first hours following the injury [4][37]. After several days, HMGB1-positive cells are mainly phagocytic microglial cells [4] (Figure 1D). Some authors have also described a biphasic expression of HMGB1 and S100 proteins with a second peak 14 days later [34][35]. The cellular consequences of DAMPs depend on the expression of PRRs but also on their post-translational modification. For instance, HMGB1 contains three highly conserved cysteines that are readily oxidized by reactive oxygen species, forming an intramolecular disulfide bridge thereby changing its conformation [41][42]. The reduced form released upon the primary injury binds the chemokine ligands (CXCL) 2 and 4 on monocytes and RAGE but not TLR-4, thereby promoting autophagy and acting as a chemoattractant for monocytes [40][43]. Conversely, the oxidized form may promote cell death and, at the late phase, vascular remodeling and progenitor cell migration via TLR signaling [35][43].

The expression of RAGE on neurons and microglial cells follows the same time course, with a biphasic pattern peaking on day 1 and 14 [12][20][34][35][36][44][45]. The amount of TLRs, which are predominantly expressed on microglial cells, increases at the subacute phase (i.e., after day 1) when activated microglia are present [34] (Figure 1D). The ligation of DAMPs to RAGE in turn activates the nuclear factor-kappa B (NF-κB) which in a positive feedback loop will increase the expression of RAGE. The activation of PRRs is related to different signaling pathways in neurons and glial cells. In neurons RAGE promotes the expression of proteins involved in necroptosis (i.e., MLKL) and autophagy (i.e., Becline-1); accordingly, blocking RAGE reduces autophagy, but also increases neuronal sensitivity to injury and apoptosis [20]. Later, at the subacute phase, TLRs and RAGE activation will polarize microglial cells and infiltrating leukocytes toward a pro-inflammatory phenotype, termed M1, with the release of cytokines such as tumor necrosis factor (TNF), IL-6 or IL-1 and the up-regulation of the inducible NO synthase [5][11][21][34][37][39]. Experimental overactivation of RAGE or TLRs by DAMPs injection or an increase in glucose degradation products, such as during hyperglycemia, will in turn increase the cytokine levels and worsen neuronal injuries [5][11][37]. The progression of secondary lesions follows different pathways. The ligation of TNF to the TNF receptor 1 (TNFR1) expressed by neurons will induce both necroptosis (via RIP1-3 and MLKL) and apoptosis (via caspase 3 and 8) [9][33]. Moreover, the pro-inflammatory cocktail can also activate astrocytes with a destructive phenotype (termed A1) leading to neuronal death and fewer synapses [7] (Figure 1C).

2.2. DAMPs Clearence

Under normal conditions, there is a continuous flow of cerebrospinal fluid (CSF) allowing the renewal of extracellular medium and the clearance of solutes such as amyloid-β (Aβ), also called the glymphatic system [46]. the subarachnoid CSF circulates in para-vascular spaces and enters the brain along penetrating arteries reaching the capillary bed; the interstitial fluid is then cleared along a perivenous drainage pathway and cervical lymphatic structures [46][47][48]. The glymphatic flux is driven by arterial pulse [47], water flux through aquaporin-4 expressed on astrocytes [46][48], as well as sleep cycles [30]. The CSF convection in the glymphatic system participates in the clearance of DAMPs to the peripheral blood [48], but also exhibits several changes following brain injury. The spreading depolarization that occurs at the very beginning of brain injuries triggers transient cell swelling, vasoconstriction [49][50] (Figure 1B), and increases the CSF flux in the glymphatic system, participating in the increase in brain water content [51]. However, the glymphatic flux is then impaired from 30 min to several days after the injury thereby leading to an accumulation of extracellular proteins such as DAMPs and Aβ [52][53][54][55]. DAMPs are also actively cleared from the extracellular space by infiltrating myeloid cells from the peripheral blood and activated resident glial cells. Infiltrating myeloid cells penetrate the lesion then differentiate into phagocytes that express scavenger receptors such as the macrophage scavenger receptor 1 (MSR1) that can bind and internalize HMGB1 and S100 proteins, as well as peridoxine [56]. These MSR1+ cells are present up to several days after the injury and exhibit an M2 phenotype, as opposed to the pro-inflammatory M1 phenotype of activated microglia that also internalize DAMPs [3][4][56]. Astrocytes are also able to internalize S100B into lysosomes by a RAGE-dependent mechanism [57]. Activated phagocytes may release HMGB1 from secretory lysosomes when modified lipids such as lysophosphatidylcholine are present at the subacute phase of acute brain injury, which may explain its biphasic release [58][59][60]. DAMPs can also traffic across the BBB by transcytosis in endothelial cells via RAGE and eventually reenter the brain from the blood compartment [61][62].

Increased expression of RAGE has been associated with the promotion of neuroinflammation in the main brain injuries, downstream HMGB1 release and NF-κB pathway activation [36]. Such a positive feedback loop of neuroinflammation may be blocked by intervention with antagonist of RAGE or the release of a truncated soluble form of RAGE (sRAGE) which may act as a decoy receptor to mitigate the inherent consequences of the inflammatory cascade. sRAGE is released upon brain injury [12], either secreted by astrocytes or monocytes [63] or from the cleavage of the membrane-bound RAGE [64]. sRAGE can scavenge extracellular and circulating DAMPs thus preventing their refilling into the brain [61] and reduce brain lesion progression [12][37][65]. The protective effect of recombinant sRAGE in cerebral parenchyma has been recently depicted in an animal model of SAH [65]. Although the characteristics of sRAGE sound interesting for clinical objectives, it has been little studied in brain injuries, with inconsistent results for outcomes such as in acute IS [66], or aSAH [31][67]. These discrepencies in data may result from different types of sRAGE studied and timing of measurements. We can speculate that the plasma level of sRAGE may be either positively associated with the intensity of injury, or negatively because of the consumption by its ligants (i.e., HMGB1 or S100B) possibly reflecting the healing process. Sometimes differences in plasma sRAGE may result from previous patient conditions such as diabetes or renal dysfunction [68]. In such a complex context it might be particularly interesting to combine more than one of these biomarker measurements to characterize the entire process.

References

  1. Kigerl, K.A.; de Rivero Vaccari, J.P.; Dietrich, W.D.; Popovich, P.G.; Keane, R.W. Pattern recognition receptors and central nervous system repair. Exp. Neurol. 2014, 258, 5–16.
  2. Kono, H.; Rock, K.L. How dying cells alert the immune system to danger. Nat. Rev. Immunol. 2008, 8, 279–289.
  3. Canton, J.; Neculai, D.; Grinstein, S. Scavenger receptors in homeostasis and immunity. Nat. Rev. Immunol. 2013, 13, 621–634.
  4. Gao, T.-L.; Yuan, X.-T.; Yang, D.; Dai, H.-L.; Wang, W.-J.; Peng, X.; Shao, H.-J.; Jin, Z.-F.; Fu, Z.-J. Expression of HMGB1 and RAGE in rat and human brains after traumatic brain injury. J. Trauma Acute Care Surg. 2012, 72, 643–649.
  5. Khan, M.A.; Schultz, S.; Othman, A.; Fleming, T.; Lebrón-Galán, R.; Rades, D.; Clemente, D.; Nawroth, P.P.; Schwaninger, M. Hyperglycemia in Stroke Impairs Polarization of Monocytes/Macrophages to a Protective Noninflammatory Cell Type. J. Neurosci. 2016, 36, 9313–9325.
  6. Bsibsi, M.; Ravid, R.; Gveric, D.; Noort, J.M. van Broad Expression of Toll-Like Receptors in the Human Central Nervous System. J. Neuropathol. Exp. Neurol. 2002, 61, 1013–1021.
  7. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487.
  8. Hu, X.; Li, P.; Guo, Y.; Wang, H.; Leak, R.K.; Chen, S.; Gao, Y.; Chen, J. Microglia/Macrophage Polarization Dynamics Reveal Novel Mechanism of Injury Expansion After Focal Cerebral Ischemia. Stroke 2012, 43, 3063–3070.
  9. Bao, Z.; Fan, L.; Zhao, L.; Xu, X.; Liu, Y.; Chao, H.; Liu, N.; You, Y.; Liu, Y.; Wang, X.; et al. Silencing of A20 Aggravates Neuronal Death and Inflammation After Traumatic Brain Injury: A Potential Trigger of Necroptosis. Front. Mol. Neurosci. 2019, 12, 222.
  10. Chaudhry, S.; Hafez, A.; Jahromi, B.; Kinfe, T.; Lamprecht, A.; Niemelä, M.; Muhammad, S. Role of Damage Associated Molecular Pattern Molecules (DAMPs) in Aneurysmal Subarachnoid Hemorrhage (aSAH). Int. J. Mol. Sci. 2018, 19, 2035.
  11. Lu, Y.; Zhang, X.-S.; Zhang, Z.-H.; Zhou, X.-M.; Gao, Y.-Y.; Liu, G.-J.; Wang, H.; Wu, L.-Y.; Li, W.; Hang, C.-H. Peroxiredoxin 2 activates microglia by interacting with Toll-like receptor 4 after subarachnoid hemorrhage. J. Neuroinflamm. 2018, 15, 87.
  12. Tang, S.-C.; Wang, Y.-C.; Li, Y.-I.; Lin, H.-C.; Manzanero, S.; Hsieh, Y.-H.; Phipps, S.; Hu, C.-J.; Chiou, H.-Y.; Huang, Y.-S.; et al. Functional Role of Soluble Receptor for Advanced Glycation End Products in Stroke. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 585–594.
  13. Pinto, A.; Tuttolomondo, A.; Raimondo, D.D.; Fernandez, P.; Licata, G. Risk factors profile and clinical outcome of ischemic stroke patients admitted in a Department of Internal Medicine and classified by TOAST classification. Int. Angiol. J. Int. Union Angiol. 2006, 25, 261–267.
  14. Shi, K.; Tian, D.-C.; Li, Z.-G.; Ducruet, A.F.; Lawton, M.T.; Shi, F.-D. Global brain inflammation in stroke. Lancet Neurol. 2019, 18, 1058–1066.
  15. Mijajlović, M.D.; Pavlović, A.; Brainin, M.; Heiss, W.-D.; Quinn, T.J.; Ihle-Hansen, H.B.; Hermann, D.M.; Assayag, E.B.; Richard, E.; Thiel, A.; et al. Post-stroke dementia—A comprehensive review. BMC Med. 2017, 15, 11.
  16. Kliper, E.; Bashat, D.B.; Bornstein, N.M.; Shenhar-Tsarfaty, S.; Hallevi, H.; Auriel, E.; Shopin, L.; Bloch, S.; Berliner, S.; Giladi, N.; et al. Cognitive Decline After Stroke. Stroke 2013, 44, 1433–1435.
  17. Dénes, Á.; Ferenczi, S.; Kovács, K.J. Systemic inflammatory challenges compromise survival after experimental stroke via augmenting brain inflammation, blood- brain barrier damage and brain oedema independently of infarct size. J. Neuroinflamm. 2011, 8, 164.
  18. Dreier, J.P.; Fabricius, M.; Ayata, C.; Sakowitz, O.W.; Shuttleworth, C.W.; Dohmen, C.; Graf, R.; Vajkoczy, P.; Helbok, R.; Suzuki, M.; et al. Recording, analysis, and interpretation of spreading depolarizations in neurointensive care: Review and recommendations of the COSBID research group. J. Cereb. Blood Flow Metab. 2017, 37, 1595–1625.
  19. Silverman, W.R.; de Rivero Vaccari, J.P.; Locovei, S.; Qiu, F.; Carlsson, S.K.; Scemes, E.; Keane, R.W.; Dahl, G. The Pannexin 1 Channel Activates the Inflammasome in Neurons and Astrocytes. J. Biol. Chem. 2009, 284, 18143–18151.
  20. Li, C.; Wu, Z.; Limnuson, K.; Cheyuo, C.; Wang, P.; Ahn, C.H.; Narayan, R.K.; Hartings, J.A. Development and application of a microfabricated multimodal neural catheter for neuroscience. Biomed. Microdevices 2016, 18, 8.
  21. Yao, X.; Liu, S.; Ding, W.; Yue, P.; Jiang, Q.; Zhao, M.; Hu, F.; Zhang, H. TLR4 signal ablation attenuated neurological deficits by regulating microglial M1/M2 phenotype after traumatic brain injury in mice. J. Neuroimmunol. 2017, 310, 38–45.
  22. Kligman, D.; Marshak, D.R. Purification and characterization of a neurite extension factor from bovine brain. Proc. Natl. Acad. Sci. USA 1985, 82, 7136–7139.
  23. Donato, R. Intracellular and extracellular roles of S100 proteins. Microsc. Res. Tech. 2003, 60, 540–551.
  24. Villarreal, A.; Seoane, R.; Torres, A.G.; Rosciszewski, G.; Angelo, M.F.; Rossi, A.; Barker, P.A.; Ramos, A.J. S100B protein activates a RAGE-dependent autocrine loop in astrocytes: Implications for its role in the propagation of reactive gliosis. J. Neurochem. 2014, 131, 190–205.
  25. Hu, Y.; Wilson, G. A temporary local energy pool coupled to neuronal activity: Fluctuations of extracellular lactate levels in rat brain monitored with rapid-response enzyme-based sensor. J. Neurochem. 1997, 69, 1484–1490.
  26. Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Othman, I.; Shaikh, M.F. HMGB1-Mediated Neuroinflammatory Responses in Brain Injuries: Potential Mechanisms and Therapeutic Opportunities. Int. J. Mol. Sci. 2020, 21, 4609.
  27. Manivannan, S.; Marei, O.; Elalfy, O.; Zaben, M. Neurogenesis after traumatic brain injury—The complex role of HMGB1 and neuroinflammation. Neuropharmacology 2021, 183, 108400.
  28. Vincent, A.M.; Perrone, L.; Sullivan, K.A.; Backus, C.; Sastry, A.M.; Lastoskie, C.; Feldman, E.L. Receptor for Advanced Glycation End Products Activation Injures Primary Sensory Neurons via Oxidative Stress. Endocrinology 2007, 148, 548–558.
  29. Kang, R.; Tang, D.; Lotze, M.T.; III, H.J.Z. RAGE regulates autophagy and apoptosis following oxidative injury. Autophagy 2011, 7, 442–444.
  30. Xie, J.; Méndez, J.D.; Méndez-Valenzuela, V.; Aguilar-Hernández, M.M. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal. 2013, 25, 2185–2197.
  31. Aida, Y.; Kamide, T.; Ishii, H.; Kitao, Y.; Uchiyama, N.; Nakada, M.; Hori, O. Soluble receptor for advanced glycation end products as a biomarker of symptomatic vasospasm in subarachnoid hemorrhage. J. Neurosurg. 2019, 1–9.
  32. Li, X.; Cheng, S.; Hu, H.; Zhang, X.; Xu, J.; Wang, R.; Zhang, P. Progranulin protects against cerebral ischemia-reperfusion (I/R) injury by inhibiting necroptosis and oxidative stress. Biochem. Bioph. Res. Commun. 2019, 521, 569–576.
  33. Croker, B.A.; Rickard, J.A.; Shlomovitz, I.; Al-Obeidi, A.; D’Cruz, A.A.; Gerlic, M. Apoptosis and Beyond; Wiley: Hoboken, NJ, USA, 2018; pp. 99–128.
  34. Yamaguchi, A.; Jitsuishi, T.; Hozumi, T.; Iwanami, J.; Kitajo, K.; Yamaguchi, H.; Mori, Y.; Mogi, M.; Sawai, S. Temporal expression profiling of DAMPs-related genes revealed the biphasic post-ischemic inflammation in the experimental stroke model. Mol. Brain 2020, 13, 57.
  35. Tian, X.; Sun, L.; Feng, D.; Sun, Q.; Dou, Y.; Liu, C.; Zhou, F.; Li, H.; Shen, H.; Wang, Z.; et al. HMGB1 promotes neurovascular remodeling via Rage in the late phase of subarachnoid hemorrhage. Brain Res. 2017, 1670, 135–145.
  36. Li, H.; Wu, W.; Sun, Q.; Liu, M.; Li, W.; Zhang, X.; Zhou, M.; Hang, C. Expression and cell distribution of receptor for advanced glycation end-products in the rat cortex following experimental subarachnoid hemorrhage. Brain Res. 2014, 1543, 315–323.
  37. Muhammad, S.; Barakat, W.; Stoyanov, S.; Murikinati, S.; Yang, H.; Tracey, K.J.; Bendszus, M.; Rossetti, G.; Nawroth, P.P.; Bierhaus, A.; et al. The HMGB1 Receptor RAGE Mediates Ischemic Brain Damage. J. Neurosci. 2008, 28, 12023–12031.
  38. Pleines, U.E.; Morganti-Kossmann, M.C.; Rancan, M.; Joller, H.; Trentz, O.; Kossmann, T. S-100 reflects the extent of injury and outcome, whereas neuronal specific enolase is a better indicator of neuroinflammation in patients with severe traumatic brain injury. J. Neurotrauma 2001, 18, 491–498.
  39. Okuma, Y.; Liu, K.; Wake, H.; Liu, R.; Nishimura, Y.; Hui, Z.; Teshigawara, K.; Haruma, J.; Yamamoto, Y.; Yamamoto, H.; et al. Glycyrrhizin inhibits traumatic brain injury by reducing HMGB1–RAGE interaction. Neuropharmacology 2014, 85, 18–26.
  40. Schiraldi, M.; Raucci, A.; Muñoz, L.M.; Livoti, E.; Celona, B.; Venereau, E.; Apuzzo, T.; Marchis, F.D.; Pedotti, M.; Bachi, A.; et al. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4The HMGB1–CXCL12 heterocomplex acts via CXCR4. J. Exp. Med. 2012, 209, 551–563.
  41. Hoppe, G.; Talcott, K.E.; Bhattacharya, S.K.; Crabb, J.W.; Sears, J.E. Molecular basis for the redox control of nuclear transport of the structural chromatin protein Hmgb1. Exp. Cell Res. 2006, 312, 3526–3538.
  42. Urbonaviciute, V.; Meister, S.; Fürnrohr, B.G.; Frey, B.; Gückel, E.; Schett, G.; Herrmann, M.; Voll, R.E. Oxidation of the alarmin high-mobility group box 1 protein (HMGB1) during apoptosis. Autoimmunity 2009, 42, 305–307.
  43. Tang, D.; Kang, R.; Cheh, C.-W.; Livesey, K.M.; Liang, X.; Schapiro, N.E.; Benschop, R.; Sparvero, L.J.; Amoscato, A.A.; Tracey, K.J.; et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 2010, 29, 5299–5310.
  44. Hassid, B.G.; Nair, M.N.; Ducruet, A.F.; Otten, M.L.; Komotar, R.J.; Pinsky, D.J.; Schmidt, A.M.; Yan, S.F.; Connolly, E.S. Neuronal RAGE expression modulates severity of injury following transient focal cerebral ischemia. J. Clin. Neurosci. 2009, 16, 302–306.
  45. Lei, C.; Zhang, S.; Cao, T.; Tao, W.; Liu, M.; Wu, B. HMGB1 may act via RAGE to promote angiogenesis in the later phase after intracerebral hemorrhage. Neuroscience 2015, 295, 39–47.
  46. Iliff, J.; Wang, M.; Liao, Y.; Plogg, B.; Peng, W.; Gundersen, G.; Benveniste, H.; Vates, G.; Deane, R.; Goldman, S.; et al. A Paravascular Pathway Facilitates CSF Flow Through the Brain Parenchyma and the Clearance of Interstitial Solutes, Including Amyloid B. Sci. Transl. Med. 2013, 4.
  47. Iliff, J.; Wang, M.; Zeppenfeld, D.; Venkataraman, A.; Plog, B.; Liao, Y.; Deane, R.; Nedergaard, M. Cerebral Arterial Pulsation Drives Paravascular CSF-Interstitial Fluid Exchange in the Murine Brain. J. Neurosci. 2019, 33.
  48. Plog, B.A.; Dashnaw, M.L.; Hitomi, E.; Peng, W.; Liao, Y.; Lou, N.; Deane, R.; Nedergaard, M. Biomarkers of Traumatic Injury Are Transported from Brain to Blood via the Glymphatic System. J. Neurosci. 2015, 35, 518–526.
  49. Risher, W.; Ard, D.; Yuan, J. Recurrent spontaneous spreading depolarizations facilitate acute dendritic injury in the ischemic penumbra. J. Neurosci. 2010, 29, 9859–9868.
  50. Dreier, J.P. The role of spreading depression, spreading depolarization and spreading ischemia in neurological disease. Nat. Med. 2011, 17, 2333.
  51. Mestre, H.; Du, T.; Sweeney, A.M.; Liu, G.; Samson, A.J.; Peng, W.; Mortensen, K.N.; Stæger, F.F.; Bork, P.A.R.; Bashford, L.; et al. Cerebrospinal fluid influx drives acute ischemic tissue swelling. Science 2020, 367, eaax7171.
  52. Iliff, J.; Chen, M.; Plog, B.; Zeppenfeld, D.; Soltero, M.; Yang, L.; Singh, I.; Deane, R.; Nedergaard, M. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J. Neurosci. 2014, 34.
  53. Gaberel, T.; Gakuba, C.; Goulay, R.; Lizarrondo, S.; Hanouz, J.-L.; Emery, E.; Touze, E.; Vivien, D.; Gauberti, M. Impaired Glymphatic Perfusion After Strokes Revealed by Contrast-Enhanced MRI. Stroke 2014, 45, 3092–3096.
  54. Goulay, R.; Flament, J.; Gauberti, M.; Naveau, M.; Pasquet, N.; Gakuba, C.; Emery, E.; Hantraye, P.; Vivien, D.; Aron-Badin, R.; et al. Subarachnoid Hemorrhage Severely Impairs Brain Parenchymal Cerebrospinal Fluid Circulation in Nonhuman Primate. Stroke 2017, 48, 2301–2305.
  55. Lindblad, C.; Nelson, D.W.; Zeiler, F.A.; Ercole, A.; Ghatan, P.H.; von Horn, H.; Risling, M.; Svensson, M.; Agoston, D.V.; Bellander, B.-M.; et al. Influence of Blood–Brain Barrier Integrity on Brain Protein Biomarker Clearance in Severe Traumatic Brain Injury: A Longitudinal Prospective Study. J. Neurotraum. 2020, 37, 1381–1391.
  56. Shichita, T.; Ito, M.; Morita, R.; Komai, K.; Noguchi, Y.; Ooboshi, H.; Koshida, R.; Takahashi, S.; Kodama, T.; Yoshimura, A. MAFB prevents excess inflammation after ischemic stroke by accelerating clearance of damage signals through MSR1. Nat. Med. 2017, 23, 723–732.
  57. Lasič, E.; Galland, F.; Vardjan, N.; Šribar, J.; Križaj, I.; Leite, M.C.; Zorec, R.; Stenovec, M. Time-dependent uptake and trafficking of vesicles capturing extracellular S100B in cultured rat astrocytes. J. Neurochem. 2016, 139, 309–323.
  58. Gardella, S.; Andrei, C.; Ferrera, D.; Lotti, L.V.; Torrisi, M.R.; Bianchi, M.E.; Rubartelli, A. The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep. 2002, 3, 995–1001.
  59. Pasvogel, A.E.; Miketova, P.; Moore, I.M. (Ki) Cerebrospinal Fluid Phospholipid Changes Following Traumatic Brain Injury. Biol. Res. Nurs. 2008, 10, 113–120.
  60. Pasvogel, A.E.; Miketova, P.; Moore, I.M. Differences in CSF Phospholipid Concentration by Traumatic Brain Injury Outcome. Biol. Res. Nurs. 2010, 11, 325–331.
  61. Deane, R.; Yan, S.D.; Submamaryan, R.K.; LaRue, B.; Jovanovic, S.; Hogg, E.; Welch, D.; Manness, L.; Lin, C.; Yu, J.; et al. RAGE mediates amyloid-β peptide transport across the blood-brain barrier and accumulation in brain. Nat. Med. 2003, 9, 907–913.
  62. Schmidt, A.M.; Hasu, M.; Popov, D.; Zhang, J.H.; Chen, J.; Yan, S.D.; Brett, J.; Cao, R.; Kuwabara, K.; Costache, G. Receptor for advanced glycation end products (AGEs) has a central role in vessel wall interactions and gene activation in response to circulating AGE proteins. Proc. Natl. Acad. Sci. 1994, 91, 8807–8811.
  63. Park, I.H.; Yeon, S.I.; Youn, J.H.; Choi, J.E.; Sasaki, N.; Choi, I.-H.; Shin, J.-S. Expression of a novel secreted splice variant of the receptor for advanced glycation end products (RAGE) in human brain astrocytes and peripheral blood mononuclear cells. Mol. Immunol. 2004, 40, 1203–1211.
  64. Raucci, A.; Cugusi, S.; Antonelli, A.; Barabino, S.M.; Monti, L.; Bierhaus, A.; Reiss, K.; Saftig, P.; Bianchi, M.E. A soluble form of the receptor for advanced glycation endproducts (RAGE) is produced by proteolytic cleavage of the membrane-bound form by the sheddase a disintegrin and metalloprotease 10 (ADAM10). Faseb J. 2008, 22, 3716–3727.
  65. Wang, W.; Lu, R.; Feng, D.; Zhang, H. Sevoflurane Inhibits Glutamate-Aspartate Transporter and Glial Fibrillary Acidic Protein Expression in Hippocampal Astrocytes of Neonatal Rats Through the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) Pathway. Anest. Analg. 2016, 123, 93.
  66. Tang, S.-C.; Yeh, S.-J.; Tsai, L.-K.; Hu, C.-J.; Lien, L.-M.; Peng, G.-S.; Yang, W.-S.; Chiou, H.-Y.; Jeng, J.-S. Cleaved but not endogenous secretory RAGE is associated with outcome in acute ischemic stroke. Neurology 2016, 86, 270–276.
  67. Yang, D.-B.; Dong, X.-Q.; Du, Q.; Yu, W.-H.; Zheng, Y.-K.; Hu, W.; Wang, K.-Y.; Chen, F.-H.; Xu, Y.-S.; Wang, Y.; et al. Clinical relevance of cleaved RAGE plasma levels as a biomarker of disease severity and functional outcome in aneurysmal subarachnoid hemorrhage. Clin. Chim. Acta 2018, 486, 335–340.
  68. Prasad, K. Low Levels of Serum Soluble Receptors for Advanced Glycation End Products, Biomarkers for Disease State: Myth or Reality. Int. J. Angiol. 2014, 23, 011–016.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 669
Revisions: 2 times (View History)
Update Date: 23 Mar 2021
1000/1000
Video Production Service