GET THE APP

Initiating Mechanisms of Surgery-induced Memory Decline: The Role
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Short Communication - (2016) Volume 7, Issue 6

Initiating Mechanisms of Surgery-induced Memory Decline: The Role of HMGB1

Susana Vacas1* and Mervyn Maze2
1Department of Anesthesia and Perioperative Medicine, University of California los Angeles, USA
2Department of Anesthesia and Perioperative Care, University of California San Francisco, USA
*Corresponding Author: Susana Vacas, Department of Anesthesia and Perioperative Medicine, University of California los Angeles, 757 Westwood Plaza, Suite 3325, Los Angeles, CA 90095, USA, Tel: +1 310-562-9407 Email:

Keywords: HMGB1; POCD; NF-κB

Introduction

We will review (i) how High Mobility Group Box Protein 1 (HMGB1) engages with the innate immune system following the aseptic trauma of surgery, (ii) how engagement of the innate immune
system results in postoperative neuroinflammation (PONI) and postoperative cognitive decline (POCD), and finally we propose (iii) putative treatments targeting these processes to limit cognitive decline.

Damage Associated Molecular Patterns and the Innate Immune System

Matzinger’s “danger” – subsequently known as “damage” - model of the immune system accounts for observations that are not explained by Burnet’s original “self-nonself ” model [1]. Necrotic cells, or cells in profound distress, release damage-associated molecular patterns (DAMPS) into the circulation. DAMPs are naturally-occurring intracellular constituents that include S100 proteins, heat-shock proteins and HMGB1. Similar to sepsis, in which release of bacterial DNA – known as pathogen-associated molecular patterns [2] – alerts the organism to invasion by “nonself ”, so too can DAMPs notify the host’s innate immune system that tissue damage is present in order to initiate healing processes. DAMPS are recognized by pattern recognition receptors (PRR) [3,4]; among PRRs, toll-like receptors (TLRs) play a key role in recognizing disparate ligands and triggering signals along different pathways. Once PRRs are ligated the immune system initiates the synthesis and release of inflammatory mediators.

High Mobility Group Box Protein 1

HMGB1 is a ubiquitous nuclear protein encoded on human chromosome 13q12-13. It was initially identified over 30 years ago with a particular focus on its importance in transcription regulation stabilizing nucleosome formation and acting as a transcription factorlike protein that regulates the expression of several genes [5], including p53, p73, the retinoblastoma family, members of the Rel/nuclear factor-κB (NF-κB) family, and nuclear hormone receptors [6]. Later studies have since recognized its cytokine-like potential [7]. HMGB1 can mediate systemic inflammatory responses by stimulating a variety of cells including monocytes, smooth muscle cells, tumor cells, dendritic cells, neutrophils and mesoangioblast stem cells, promoting inflammation, angiogenesis, and enhancing hematopoietic stem-cell migration [8]. Emerging evidence indicates that HMGB1 is also an essential mediator of severe systemic inflammation (SSI) [9,10].

HMGB1 can be released into the extracellular space either by an active process, once it is acetylated in the nucleus, or by a passive process in which it diffuses out of the necrotic cells. Various stimuli can activate monocytes, tissue macrophages, NK or even mature myeloid dendritic cells to secrete HMGB1 [5]. Depending on the ambient redox state the cysteine residues of HMGB1 may become oxidized thereby dictating the subsequent signaling [11,12]. HMGB1 contains three conserved cysteines that are sensitive to oxidation: Cys23, Cys45 (located in Box A), and Cys106 (located in Box B) [12]. Cys-23 and Cys-45 can easily form a disulfide bond under relatively mild oxidative conditions giving rise to “disulfide HMGB1”. Cys-106 remains in the reduced state until it is exposed to a large amount of reactive oxygen species, typically from activated leukocytes, thereby forming “sulfonated HMGB1” [6]. When released into the extracellular space, HMGB1 is initially in the reduced state but becomes oxidized by the extant environmental conditions that initiated its release. The disulfide form of HMGB1 binds to TLR2, TLR4 and receptor for advanced glycation end-products (RAGE). Signaling through these receptors leads to activation of NF-κB, that is pivotal for the proinflammatory pathway, thereby increasing the synthesis and release of cytokines [13]. As triggering of PRRs on inflammatory cells by disulfide HMGB1 generates more reactive oxygen species, further oxidation generates sulfonated HMGB1 thereby resolving inflammation [14]. Interestingly, while extracellular disulfide HMGB1 leads to the synthesis and release of tumor necrosis factor (TNF)α, intracellular HMGB1 may silence TNFα during SSI, as seen in sepsis and other disseminated acute inflammatory processes [15]; evidence for this role comes from studies involving the depletion of HMGB1 in which RelB dissociates from the promoter region and negatively regulates the expression of TNF-α [16]. In addition, loading of HMGB1 at the TNF-α gene locus correlates with the presence of H3K9me2 [15], which implies that histone methyltransferase G9a and its partner protein GLP may also play roles in the regulation of SSI [17]. Recent observations indicate that the G9a/GLP complex is also involved in the maintenance of imprinted DNA methylation [18], the potential epigenetic mechanisms associated with SSI may also include DNA methylation.

Postoperative Neuroinflammation and Postoperative Cognitive Decline (Figure 1)

The discharge of proinflammatory cytokines into the circulation is responsible for the disruption of the blood brain barrier (BBB) integrity [19]. The pathways that lead to neuroinflammation and cognitive decline after infection seem to be different from those following aseptic surgical trauma [20]. After aseptic peripheral surgery, the release of monocyte chemotactic protein-1 (MCP-1) in the hippocampus is responsible for attracting chemokine C-C motif receptor 2-expressing cells into the brain [21]. In the active state, the translocated monocytes, now macrophages, synthesize and release a variety of pro-inflammatory cytokines that are capable of disrupting long-term potentiation, the neurobiologic correlate of memory. Depletion of bone marrow-derived macrophages (BM-DM) reduces, PONI and POCD [21]. Interestingly, depletion of BM-DM does not affect surgery-induced upregulation of hippocampal MCP-1 suggesting that it originates from cells other than BM-DM, mostly likely activated microglia [8,22]. Chronic neuroinflammation with persistently activated microglia in the hippocampus can disrupt neuronal networks required for learning and memory [23].

clinical-cellular-immunology-cognitive-dysfunction

Figure 1: Represents the pathways from peripheral trauma to cognitive dysfunction.

N-Methyl-D-aspartate (NMDA) receptors are highly concentrated in the hippocampus, playing an important role in neuroplasticity and long-term potentiation is crucially dependent on these receptors [24,25]; PONI can lead to a reduction in the number of these receptors [26]. During early stages of neuroinflammation dysfunction at the NMDA-glutamate synapse may disrupt accuracy of information processing, while in the later stages of neuroinflammation it may progress to neuronal degeneration [27,28].

Putative, additional, mechanisms whereby proinflammatory cytokines induce a long-lasting effect on cognitive function may be due to disruption of specific neural circuits [29], neurogenesis [30] as well as epigenetic modifications [31].

Possible Targets for Therapeutic Interventions of Postoperative Cognitive Decline

Changes in subcortical white matter [31], brain plasticity [32], as well as information-processing circuits within the hippocampus are features related to aging [33]. Further, declines in immune function [34] within this population lead to increased neuroinflammation and coincide with cognitive decline [35], making this patient population particularly vulnerable to POCD. With an ever-aging surgical population, postoperative delirium (POD), an easily diagnosed form of POCD, has reached epidemic proportions in the US. According to National Hospital Survey Data for 2010 more than 19 million inpatients over the age of 65% had surgery. With a quoted incidence of POD in this age group of between 15-53% [32], this translates into up to 10 million POD sufferers each year. While POD is usually shortlived, its occurrence may presage long-term problems that are associated with longer hospital stays, higher morbidity and mortality, higher risks of institutionalization, cognitive decline/dementia, and poorer overall outcomes [33-36]. Average health care costs per day survived among patients with delirium have been estimated to be more than 2.5 times those of patients without delirium, leading to increased cost of care of between $38−152 billion/year [37].

Beyond the numbers that describe the scale of the problem and its healthcare costs is the immense burden that POD places on the patient for whom “staying sharp” was noted to be the most highly desired commodity with aging [38]. For the patient’s care-giver, apart from the distress of watching their beloved’s cognitive deterioration is the need to provide alternatives to overcome the patient’s deficits in activities of daily living.

For these reasons therapeutic interventions are urgently required to address this serious condition. Tracking the precise biological and cellular pathways involved in postoperative memory deficits are decisive components in mapping appropriate interventions to prevent this postoperative complication. The detection of HMGB involvement in the initiation of the surgery-induced PONI, provides an opportunity for manipulating the function of HMGB1 and modifying DAMP signaling. Pre-surgical administration of HMGB1 antibody is able to halt the neuroinflammatory response to surgery by attenuating MCP-1 production [8]. Further studies are needed to determine whether this form of immunotherapy represents a viable option for preventing POD.

Acknowledgments

Mervyn Maze has received support from NIH grant R01GM104194.

References

  1. Matzinger P (2002) The danger model: a renewed sense of self. Science 296: 301-305.
  2. Lotze M (2007) The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev 220: 60-81.
  3. Zhang Q (2010) Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464: 104-107.
  4. Rosczyk HA, Sparkman NL, Johnson RW (2008) Neuroinflammation and cognitive function in aged mice following minor surgery. Exp Gerontol 4: 840-846.
  5. Lotze MT, Tracey KJ (2005) High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 5: 331-342.
  6. Tang D, Kang R, Zeh HJ, Lotze MT (2011) High-mobility group box, oxidative stress, and disease. Antioxid Redox Signal 14: 1315-1335.
  7. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, et al. (1999) HMG-1 as a late mediator of endotoxin lethality in mice. Science 285: 248-251.
  8. Vacas S, Degos V, Tracey KJ, Maze M (2013) High-mobility Group Box 1 Protein Initiates Postoperative Cognitive Decline by Engaging Bone Marrow-derived Macrophages. Anesthesiology.
  9. Müller S, Scaffidi P, Degryse B, Bonaldi T, Ronfani L, et al. (2001) New EMBO members' review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J 20: 4337-4340.
  10. Wang H, Liao H, Ochani M, Justiniani M, Lin X, et al. (2004) Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat Med 10: 1216-1221.
  11. Carta S, Castellani P, Delfino L, Tassi S, Venè R, et al. (2009) DAMPs and inflammatory processes: the role of redox in the different outcomes. J Leukoc Biol 86: 549-555.
  12. Janko C, Filipovia M, Munoz LE, Schorn C, Schett G, et al. (2014) Redox modulation of HMGB1-related signaling. Antioxid Redox Signal 20: 1075-1085.
  13. Tracey KJ (2009) Reflex control of immunity. Nat Rev Immunol 9: 418-428.
  14. Zandarashvili L (2013) Real-time kinetics of high-mobility group box 1 (HMGB1) oxidation in extracellular fluids studied by in situ protein NMR spectroscopy. J Biol Chem 288: 11621-11627.
  15. El Gazzar M, Yoza BK, Chen X, Garcia BA, Young NL, et al. (2009) Chromatin-specific remodeling by HMGB1 and linker histone H1 silences proinflammatory genes during endotoxin tolerance. Mol Cell Biol 29: 1959-1971.
  16. Andersson U, Wang H, Palmblad K, Aveberger AC, Bloom O, et al. (2000) High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J Exp Med 192: 565-570.
  17. Tachibana M, Sugimoto K, Fukushima T, Shinkai Y (2001) Set domain-containing protein, G9a, is a novel lysine-preferring mammalian histone methyltransferase with hyperactivity and specific selectivity to lysines 9 and 27 of histone H3. J Biol Chem 276: 25309-25317.
  18. Zhang T, Termanis A, Özkan B, Bao XX, Culley J, et al. (2016) G9a/GLP Complex Maintains Imprinted DNA Methylation in Embryonic Stem Cells. Cell Rep 15: 77-85.
  19. Terrando N, Eriksson LI, Ryu JK, Yang T, Monaco C, et al. (2011) Resolving postoperative neuroinflammation and cognitive decline. Ann Neurol 70: 986-995.
  20. Terrando N, Monaco C, Ma D, Foxwell BM, Feldmann M, et al. (2010) Tumor necrosis factor-alpha triggers a cytokine cascade yielding postoperative cognitive decline. Proc Natl Acad Sci U S A 107: 20518-20522.
  21. Degos V, Vacas S, Han Z, van Rooijen N, Gressens P, et al. (2013) Depletion of Bone Marrow-derived Macrophages Perturbs the Innate Immune Response to Surgery and Reduces Postoperative Memory Dysfunction. Anesthesiology 118: 527-536.
  22. Zhang X, Dong H, Li N, Zhang S, Sun J, et al. (2016) Activated brain mast cells contribute to postoperative cognitive dysfunction by evoking microglia activation and neuronal apoptosis. J. Neuroinflammation 1: 127.
  23. Sajja VSSS, Hlavac N, VandeVord PJ (2016) Role of Glia in Memory Deficits Following Traumatic Brain Injury: Biomarkers of Glia Dysfunction. Front Integr Neurosci 10: 7.
  24. Bauer EP, Schafe GE, LeDoux JE (2002) NMDA Receptors and L-Type Voltage-Gated Calcium Channels Contribute to Long-Term Potentiation and Different Components of Fear Memory Formation in the Lateral Amygdala. J Neurosci 2.
  25. Berberich S, Jensen V, Hvalby Ø, Seeburg PH, Köhr G (2007) The role of NMDAR subtypes and charge transfer during hippocampal LTP induction. Neuropharmacology 5: 77-86.
  26. Zhang X, Xin X, Dong Y, Zhang Y, Yu B, et al. (2013) Surgical incision-induced nociception causes cognitive impairment and reduction in synaptic NMDA receptor 2B in mice. J Neurosci 3: 17737-17748.
  27. Rosi S, Ramirez-Amaya V, Hauss-Wegrzyniak B, Wenk GL (2004) Chronic brain inflammation leads to a decline in hippocampal NMDA-R1 receptors. J Neuroinflammation 1: 12.
  28. Rosi S, Ramirez-Amaya V, Vazdarjanova A, Esparza EE, Larkin PB, et al. (2009) Accuracy of hippocampal network activity is disrupted by neuroinflammation: rescue by memantine. Brain 132: 2464-2477.
  29. Czerniawski J, Guzowski JF (2014) Acute Neuroinflammation Impairs Context Discrimination Memory and Disrupts Pattern Separation Processes in Hippocampus. J Neurosci 34.
  30. de Miranda AS, Zhang CJ, Katsumoto A, Teixeira AL (2016) Hippocampal adult neurogenesis: Does the immune system matter? J Neurol Sci.
  31. Donzis EJ, Tronson NC (2014) Modulation of learning and memory by cytokines: signaling mechanisms and long term consequences. Neurobiol Learn Mem 115: 68-77.
  32. Inouye SK, Westendorp RGJ, Saczynski JS (2014) Delirium in elderly people. Lancet (London, England) 38: 911-922.
  33. Sanders RD, Pandharipande PP, Davidson AJ, Ma D, Maze M (2011) Anticipating and managing postoperative delirium and cognitive decline in adults. BMJ 343: d4331.
  34. Fong TG, Jones RN, Shi P, Marcantonio ER, Yap L, et al. (2009) Delirium accelerates cognitive decline in Alzheimer disease. Neurology 72: 1570-1575.
  35. Fong TG, Tulebaev SR, Inouye SK (2009) Delirium in elderly adults: diagnosis, prevention and treatment. Nat Rev Neurol 5: 210-220.
  36. Inouye SK (2006) Delirium in older persons. N Engl J Med 354: 1157-1165.
  37. Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK (2008) One-year health care costs associated with delirium in the elderly population. Arch Intern Med 168: 27-32.
  38. Sachs-Ericsson N, Blazer DG (2015) The new DSM-5 diagnosis of mild neurocognitive disorder and its relation to research in mild cognitive impairment. Aging Ment Health 19: 2-12.
Citation: Vacas S, Maze M (2016) Initiating Mechanisms of Surgery-induced Memory Decline: The Role of HMGB1. J Clin Cell Immunol 7:481.

Copyright: © 2016 Vacas S, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top